1
|
Xiao S, Shen Y, Zhang M, Liu X, Cai T, Wang F. VacA promotes pyroptosis via TNFAIP3/TRAF1 signaling to induce onset of atrophic gastritis. Microbiol Res 2025; 296:128142. [PMID: 40138873 DOI: 10.1016/j.micres.2025.128142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Atrophic gastritis (AG) is a chronic inflammation where gastric glandular cells are replaced by intestinal-type epithelium. Gastric epithelial cell loss is often linked to multiple cell death signaling pathways. While Helicobacter pylori (H. pylori) infection is the main cause of AG, its role in inducing cell death goes beyond apoptosis and autophagy. Pyroptosis could promote development of inflammation related cancers, but its involvement in H. pylori-induced malignant transformation remains unclear. METHODS The enrichment of pyroptosis signaling across pathological stages was assessed using immunohistochemistry and bioinformatic analysis. Gastric epithelial cells were co-cultured with VacA recombinant protein or VacA+H. pylori to investigate the role of VacA in pyroptosis, and its downstream targets. TNFAIP3 or TRAF1 was silenced/overexpressed in gastric epithelial cells to explore their impact on pyroptosis. Finally, the interaction between TNFAIP3 and TRAF1 was examined using Western Blot, immunofluorescence, co-immunoprecipitation and ubiquitin assays. RESULTS Expression of pyroptosis components and pyroptosis enrichment score were upregulated in AG and gastric cancer tissues compared to normal or non-atrophic gastritis tissues. Upon incubation with VacA recombinant protein or VacA+H. pylori, pyroptosis and TNFAIP3/TRAF1 were elevated in gastric epithelial cells. TRAF1 promoted expression of downstream pyroptosis components and release of IL-1β/IL18. TRAF1 ablation could reverse pyroptosis activation caused by VacA. Finally, we proved TNFAIP3 as deubiquitinating enzyme to increase TRAF1 stability, further inducing pyroptosis. CONCLUSIONS The VacA/TNFAIP3/TRAF1 signaling cascade facilitates pyroptosis in H. pylori- infected tissue. Overactivation of Pyroptosis caused the atrophy-like morphological changes of gastric epithelium, further inducing sustainable malignant transformation.
Collapse
Affiliation(s)
- Shilang Xiao
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China
| | - Yicun Shen
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China
| | - Minglin Zhang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China
| | - Xiaoming Liu
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China.
| | - Ting Cai
- Department of gastroenterology, Hunan provincial people's hospital, the first affiliated hospital of Hunan Normal University, 61 Jiefang Road, Changsha, Hunan 410005, China.
| | - Fen Wang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China.
| |
Collapse
|
2
|
Li Y, Zhou B, Liu D, Nie G, Yang F, Chen J, Cheng S, Kang Y, Liu B, Dong B, Liu M. Carbon monoxide gas molecules: Therapeutic mechanisms in radiation-induced lung injury. J Colloid Interface Sci 2025; 688:250-263. [PMID: 40010090 DOI: 10.1016/j.jcis.2025.02.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Radiation therapy (RT) remains an essential treatment modality for lung cancer, yet its effectiveness is frequently hindered by radiation-induced lung injury (RILI), a common outcome of modern therapeutic regimens. With the aim of addressing this challenge, a novel nanocomposite, Au@mSiO2@Mn(CO)5Br (ASMB), was synthesized with Au@mSiO2 as the carrier and Mn(CO)5Br as the functional component. The gold nanorods (Au rods) core generates reactive oxygen species (ROS) under X-ray irradiation, which then activates Mn(CO)5Br to release carbon monoxide (CO) locally within the lung during radiotherapy. The released CO then diffuses to surrounding tissues, inhibiting the excessive accumulation of ROS, thereby preventing damage to normal cells caused by ROS generated in a short period of time. Meanwhile, the released manganese ions (Mnn+) catalyze the conversion of hydrogen peroxide (H2O2) in the microenvironment into oxygen (O2). In vitro experiments demonstrated that the release of CO markedly attenuated radiation-induced ROS production, thereby inhibiting the activation of the NLRP3 inflammasome and reducing the levels of inflammatory cytokines and pyroptosis-related proteins. Moreover, it downregulated the expression of fibrosis-associated proteins, including TGF-β1 and α-SMA. Additionally, CO facilitated DNA damage repair, thereby mitigating radiation-induced tissue injury. In the RILI model, the ASMB NPs-treated lungs exhibited notably reduced pulmonary edema, congestion, and inflammatory cell infiltration, primarily by inhibiting NLRP3 inflammasome-dependent pyroptosis and reducing levels of inflammation and fibrosis markers. The release of O2 further mitigates local tissue hypoxia, enhancing the effectiveness of radiotherapy. Overall, ASMB NPs provide a promising alternative for the treatment of RILI and a potential therapeutic strategy to improve the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Ya'nan Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Bingshuai Zhou
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130021, China
| | - Dajie Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Guodong Nie
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Fan Yang
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Jiajie Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Sen Cheng
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Yahui Kang
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China; Department of Radiation Oncology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230031, China
| | - Bailong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China.
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130021, China.
| | - Min Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China.
| |
Collapse
|
3
|
Atabay M, Inci F, Saylan Y. Computational studies for the development of extracellular vesicle-based biosensors. Biosens Bioelectron 2025; 277:117275. [PMID: 39999607 DOI: 10.1016/j.bios.2025.117275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/25/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Cancer affects millions of people, and early detection and efficient treatment are two strong levers to hurdle this disease. Recent studies on exosomes, a subset of extracellular vesicles, have deliberately shown the potential to function as a biomarker or treatment tool, thereby attracting the attention of researchers who work on developing biosensors. Due to the ability of computational methods to predict of the behavior of biomolecules, the combination of experimental and computational methods would enhance the analytical performance of the biosensor, including sensitivity, accuracy, and specificity, even detecting such vesicles from bodily fluids. In this regard, the role of computational methods such as molecular docking, molecular dynamics simulation, and density functional theory is overviewed in the development of biosensors. This review highlights the investigations and studies that have been reported using these methods to design exosome-based biosensors. This review concludes with the role of the quantum mechanics/molecular mechanics method in the investigation of chemical processes of biomolecular systems and the deficiencies in using this approach to develop exosome-based biosensors. In addition, the artificial intelligence theory is explained briefly to show its importance in the study of these biosensors.
Collapse
Affiliation(s)
- Maryam Atabay
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, Turkey; Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Yeşeren Saylan
- Department of Chemistry, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
4
|
Wu X, Pan Q, Yao C, Gong Y, Li Z, Tang F, Fang Z, Bao Y, Chen Y, Yu M, Wang Z, Jiang C, Hong Z. Therapeutic potential of quercitrin in intervertebral disc degeneration: Targeting pyroptosis and inflammation. Int Immunopharmacol 2025; 156:114680. [PMID: 40273673 DOI: 10.1016/j.intimp.2025.114680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 03/20/2025] [Accepted: 04/13/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Intervertebral disc degeneration (IDD) is the predominant cause of low back pain (LBP), that leads to significant disability and imposes a substantial socioeconomic burden. Despite its prevalence, effective treatment for IDD has yet to be fully established. This study aimed to explore the therapeutic potential of quercitrin (QUE) in IDD development and to elucidate its underlying mechanisms. METHODS In vitro, we investigated the effects of QUE on ECM metabolism, inflammatory response and pyroptosis in IL-1β-stimulated nucleus pulposus cells (NPCs), along with the potential mechanisms. In vivo, mice lumbar spinal instability (LSI) was established to determined the impact of QUE on IDD progression. RESULTS QUE significantly alleviated inflammation and maintained the ECM homeostasis under IL-1β stimulation in NPCs. Moreover, QUE appeared to inhibit pyroptosis, which was closely related to intense inflammatory response. Notably, the protective effects of QUE were abrogated upon inhibition of TRIM31 activity, indicating that TRIM31 mediated pyroptosis suppression is crucial for the therapeutic effects of QUE. CONCLUSION QUE plays an important role in alleviating pyroptosis and inflammation within NPCs, thereby slowing the progression of IDD. Therefore, QUE might emerge as a promising therapeutic candidate for IDD, holding the potential for clinical application in the future.
Collapse
Affiliation(s)
- Xinyu Wu
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Qiaohong Pan
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Can Yao
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Yuhang Gong
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Ze Li
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Fang Tang
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Zhiyu Fang
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Yuxuan Bao
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Yiyu Chen
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Minyang Yu
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China
| | - Zhangfu Wang
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China.
| | - Chao Jiang
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China.
| | - Zhenghua Hong
- Department of Orthopaedics, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang Province, China; Bone Development and Metabolism Research Center of Taizhou Hospital, Zhejiang Province, Linhai, Zhejiang Province, China.
| |
Collapse
|
5
|
Wu S, Luo Y, Wei F, Li Y, Fan J, Chen Y, Zhang W, Li X, Xu Y, Chen Z, Xia C, Hu M, Li P, Gu Q. Lactic acid bacteria target NF-κB signaling to alleviate gastric inflammation. Food Funct 2025; 16:3101-3119. [PMID: 40152095 DOI: 10.1039/d4fo06308b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Helicobacter pylori (H. pylori) infection and the resulting gastric inflammation are major contributors to gastric cancer development. Probiotics, particularly Lactobacillus, are promising for their anti-inflammatory potential, yet their exact mechanisms in inhibiting H. pylori-induced inflammation are unclear. In our previous study, Lactiplantibacillus plantarum ZJ316 (L. plantarum ZJ316) demonstrated strong anti-inflammatory effects against H. pylori infection in vivo, but its precise mechanisms were not fully understood. Here, we aimed to investigate how L. plantarum ZJ316 inhibits the inflammatory response to H. pylori infection. Our results demonstrated that L. plantarum ZJ316 effectively reduced the expression of pro-inflammatory cytokines in H. pylori-infected AGS cells. Mechanistically, L. plantarum ZJ316 inhibited the NF-κB signaling pathway by preventing the degradation of IκBα, suppressing p65 phosphorylation, and blocking the nuclear translocation of phosphorylated p65. Treatment with the NF-κB inhibitor BAY 11-7082 further decreased tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8), and interleukin-1β (IL-1β) levels, confirming the inhibitory effect of L. plantarum ZJ316 on the NF-κB pathway. In H. pylori-infected mice, oral administration of L. plantarum ZJ316 significantly alleviated inflammatory cell infiltration, reduced TNF-α and pepsinogen II (PGII) levels, and increased interleukin-10 (IL-10) levels in serum. A comparative metagenomic analysis of the gastric microbiota revealed a decrease in Prevotella and Desulfovibrio, alongside an increase in Ligilactobacillus and Akkermansia, supporting the protective effects of L. plantarum ZJ316 and correlating with their decreased inflammatory response. In summary, administration of L. plantarum ZJ316 demonstrated robust anti-inflammatory effects against H. pylori infection by suppressing NF-κB signaling and promoting favorable changes in the gastric microbiota composition. Therefore, L. plantarum ZJ316 holds promise as a novel functional food for protecting the body against H. pylori infection.
Collapse
Affiliation(s)
- Shiying Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yuenuo Luo
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Fangtong Wei
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Jiayi Fan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yongqiang Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Wenjie Zhang
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Xuelong Li
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Yang Xu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ziqi Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Chenlan Xia
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Mingyang Hu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
6
|
Carnazzo V, Rigante D, Restante G, Basile V, Pocino K, Basile U. The entrenchment of NLRP3 inflammasomes in autoimmune disease-related inflammation. Autoimmun Rev 2025; 24:103815. [PMID: 40233890 DOI: 10.1016/j.autrev.2025.103815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Autoinflammation and autoimmunity are almost "opposite" phenomena characterized by chronic activation of the immune system, 'innate' in the first and 'adaptive' in the second, leading to inflammation of several tissues with specific protean effectors of tissue damage. The mechanism of involvement of multiprotein complexes called 'inflammasomes' within autoimmune pictures, differently from autoinflammatory conditions, is yet undeciphered. In this review we provide a comprehensive overview on NLRP3 inflammasome contribution into the pathogenesis of some autoimmune diseases. In response to autoantibodies against nucleic acids or tissue-specific antigens the NLRP3 inflammasome is activated within dendritic cells and macrophages of patients with systemic lupus erythematosus. Crucial is NLRP3 inflammasome to amplify tissue inflammation with interleukin-1 overexpression and matrix metalloproteinase production at the joint level in rheumatoid arthritis. A deregulated NLRP3 inflammasome activation occurs in the serous acini of salivary and lacrimal glands prone to Sjogren's syndrome, but also in the inflammatory process involving endothelial cells, leucocyte recruitment, and platelet plugging of vasculitides. Furthermore, organ-specific autoimmune diseases such as thyroiditis and hepatitis may display hyperactive NLRP3 inflammasomes at the level of resident immune cells within thyroid or liver, respectively. Therefore, it is not unexpected that preclinical studies have shown how specific inflammasome inhibitors may significantly overthrow the severity of different autoimmune diseases and slow down their trend towards an ominous progression. Specific markers of inflammasome activation could also reveal subclinical inflammatory components escaping conventional diagnostic approaches or improve monitoring of autoimmune diseases and personalizing their treatment.
Collapse
Affiliation(s)
- Valeria Carnazzo
- Department of Clinical Pathology, Santa Maria Goretti Hospital, Latina, Italy.
| | - Donato Rigante
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica Sacro Cuore, Rome, Italy.
| | - Giuliana Restante
- Department of Experimental Medicine, University "La Sapienza", Rome, Italy
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Krizia Pocino
- Unit of Clinical Pathology, Ospedale San Pietro Fatebenefratelli, Rome, Italy
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti Hospital, Latina, Italy.
| |
Collapse
|
7
|
Wu J, Yang Z, Chen X, Hou S, Li N, Chang Y, Yin J, Xu J. TRIM36 Inhibits the Development of AOM/DSS-Induced Colitis-Associated Colorectal Cancer by Promoting the Ubiquitination and Degradation of GRB7. Mol Carcinog 2025; 64:668-679. [PMID: 39803720 DOI: 10.1002/mc.23871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/30/2024] [Accepted: 12/05/2024] [Indexed: 03/10/2025]
Abstract
Colorectal cancer (CRC) is among the most common cancer types for both sexes. Tripartite motif 36 (TRIM36) has been reported to be aberrantly expressed in several cancer types, suggesting its involvement in cancer progression. However, the role of TRIM36 in the colorectal carcinogenesis remain unknown. In our in vivo experiments, we investigated the role of TRIM36 in AOM/DSS-induced colitis-associated carcinogenesis using TRIM36-knockout (TRIM36 KO) mice. Subsequently, we overexpressed and knocked down TRIM36 expression in two CRC cell lines to further confirm the role of TRIM36 in vitro. The UALCAN database revealed a significant decrease in TRIM36 levels in CRC tissues, including colon adenocarcinoma and rectum adenocarcinoma. A significant correlation was observed between TRIM36 levels and the histological subtype, individual cancer stage, and nodal metastasis status. The downregulation of TRIM36 in CRC tissues was further confirmed using our own collected clinical specimens. Low expression of TRIM36 was found to be associated with unfavorable overall survival and recurrence-free survival in CRC. TRIM36 KO promoted inflammation, inhibited autophagy, and facilitated the development of AOM/DSS-induced CRC. TRIM36 overexpression inhibited proliferation, migration, and invasion, while activated autophagy in CRC cells. TRIM36 directly bound to and regulated the ubiquitination of GRB7 protein. The tumor-suppressive role of TRIM36 in CRC cells was mediated by GRB7. The TRIM36/GRB7 axis may represent a promising therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Ju Wu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Zhengbo Yang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Xi Chen
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Shuangshuang Hou
- Department of General Surgery, Fuyang Normal University Second Affiliated Hospital, Fuyang, China
| | - Nanbo Li
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yaoyuan Chang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Jiajun Yin
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Jian Xu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| |
Collapse
|
8
|
Mohapatra B, Lavudi K, Kokkanti RR, Patnaik S. Regulation of NLRP3/TRIM family signaling in gut inflammation and colorectal cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189271. [PMID: 39864469 DOI: 10.1016/j.bbcan.2025.189271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
CRC (Colorectal cancer) ranks among the most prevalent tumors in humans and remains a leading cause of cancer-related mortality worldwide. Numerous studies have highlighted the connection between inflammasome over-activation and the initiation and progression of CRC. The activation of the NLRP3 (NOD-like receptor family, pyrin domain containing 3) inflammasome is dependent on the nuclear NF-kβ (Nuclear Factor kappa-light-chain-enhancer of activated B cells) pathway, leading to the maturation and release of inflammatory cytokines such as IL-1ß (Interleukin 1 beta) and IL-18 (Interleukin 18). While inflammation is crucial for defense mechanisms and tissue repair, excessive information can pose significant risks. Mounting evidence suggests that overactivation of the inflammasome contributes to the pathogenesis of inflammatory diseases. Consequently, there is a concerted effort to tightly regulate inflammasome activity and mitigate excessive inflammatory responses, particularly in conditions such as IBD (Inflammatory Bowel Disease), which includes Ulcerative Colitis and Crohn's Disease. The tripartite motif (TRIM) protein family, characterized by a conserved structure and rapid evolutionary diversification, includes members with critical roles in ubiquitination and other regulatory functions. Their importance in modulating inflammatory responses is widely acknowledged. This article aims to investigate the interplay between TRIM proteins and the NLRP3 Inflammasome in CRC and gut inflammation, offering insights for future research endeavors and potential therapeutic strategies.
Collapse
Affiliation(s)
- Bibhashee Mohapatra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Kousalya Lavudi
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, United States; Comprehensive cancer center, The Ohio State University, Columbus, OH, United States
| | - Rekha Rani Kokkanti
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Srinivas Patnaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
9
|
Beesetti S. Ubiquitin Ligases in Control: Regulating NLRP3 Inflammasome Activation. FRONT BIOSCI-LANDMRK 2025; 30:25970. [PMID: 40152367 DOI: 10.31083/fbl25970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 03/29/2025]
Abstract
Ubiquitin ligases play pivotal roles in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, a critical process in innate immunity and inflammatory responses. This review explores the intricate mechanisms by which various E3 ubiquitin ligases exert both positive and negative influences on NLRP3 inflammasome activity through diverse post-translational modifications. Negative regulation of NLRP3 inflammasome assembly is mediated by several E3 ligases, including F-box and leucine-rich repeat protein 2 (FBXL2), tripartite motif-containing protein 31 (TRIM31), and Casitas B-lineage lymphoma b (Cbl-b), which induce K48-linked ubiquitination of NLRP3, targeting it for proteasomal degradation. Membrane-associated RING-CH 7 (MARCH7) similarly promotes K48-linked ubiquitination leading to autophagic degradation, while RING finger protein (RNF125) induces K63-linked ubiquitination to modulate NLRP3 function. Ariadne homolog 2 (ARIH2) targets the nucleotide-binding domain (NBD) domain of NLRP3, inhibiting its activation, and tripartite motif-containing protein (TRIM65) employs dual K48 and K63-linked ubiquitination to suppress inflammasome assembly. Conversely, Pellino2 exemplifies a positive regulator, promoting NLRP3 inflammasome activation through K63-linked ubiquitination. Additionally, ubiquitin ligases influence other components critical for inflammasome function. TNF receptor-associated factor 3 (TRAF3) mediates K63 polyubiquitination of apoptosis-associated speck-like protein containing a CARD (ASC), facilitating its degradation, while E3 ligases regulate caspase-1 activation and DEAH-box helicase 33 (DHX33)-NLRP3 complex formation through specific ubiquitination events. Beyond direct inflammasome regulation, ubiquitin ligases impact broader innate immune signaling pathways, modulating pattern-recognition receptor responses and dendritic cell maturation. Furthermore, they intricately control NOD1/NOD2 signaling through K63-linked polyubiquitination of receptor-interacting protein 2 (RIP2), crucial for nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) activation. Furthermore, we explore how various pathogens, including bacteria, viruses, and parasites, have evolved sophisticated strategies to hijack the host ubiquitination machinery, manipulating NLRP3 inflammasome activation to evade immune responses. This comprehensive analysis provides insights into the molecular mechanisms underlying inflammasome regulation and their implications for inflammatory diseases, offering potential avenues for therapeutic interventions targeting the NLRP3 inflammasome. In conclusion, ubiquitin ligases emerge as key regulators of NLRP3 inflammasome activation, exhibiting a complex array of functions that finely tune immune responses. Understanding these regulatory mechanisms not only sheds light on fundamental aspects of inflammation but also offers potential therapeutic avenues for inflammatory disorders and infectious diseases.
Collapse
Affiliation(s)
- Swarna Beesetti
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
10
|
Wang H, Ma L, Su W, Liu Y, Xie N, Liu J. NLRP3 inflammasome in health and disease (Review). Int J Mol Med 2025; 55:48. [PMID: 39930811 PMCID: PMC11781521 DOI: 10.3892/ijmm.2025.5489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Activation of inflammasomes is the activation of inflammation‑related caspase mediated by the assembly signal of multi‑protein complex and the maturity of inflammatory factors, such as IL‑1β and IL‑18. Among them, the Nod‑like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most thoroughly studied type of inflammatory corpuscle at present, which is involved in the occurrence and development of numerous human diseases. Therefore, targeting the NLRP3 inflammasome has become the focus of drug development for related diseases. In this paper, the research progress of the NLRP3 inflammasome in recent years is summarized, including the activation and regulation of NLRP3 and its association with diseases. A deep understanding of the regulatory mechanism of NLRP3 will be helpful to the discovery of new drug targets and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Weiran Su
- Department of Internal Medicine, Jiading District Central Hospital, Shanghai 201800, P.R. China
| | - Yangruoyu Liu
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ning Xie
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
11
|
Ma Y, Jia R, Chen S, Ma J, Yin L, Pan X, He Y, Wu T, Zhao Z, Ma L, Wu S, Wang H, Liang G, Huang S, Sun X. Ubiquitin-Proteasome System in Periodontitis: Mechanisms and Clinical Implications. Cell Prolif 2025; 58:e13781. [PMID: 39626954 PMCID: PMC11882760 DOI: 10.1111/cpr.13781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/18/2024] [Accepted: 11/09/2024] [Indexed: 01/03/2025] Open
Abstract
The progression of periodontitis, a bacteria-driven inflammatory and bone-destructive disease, involves myriad cellular and molecular mechanisms. Protein regulation significantly influences the pathogenesis and management of periodontitis. However, research regarding its regulatory role in periodontitis remains relatively limited. The ubiquitin-proteasome system (UPS), which mainly involves ubiquitination by E3 ubiquitin ligases (E3s) and deubiquitination by deubiquitinating enzymes (DUBs), is the primary intracellular and non-lysosomal mechanism of protein degradation. Recent studies have provided compelling evidence to support the involvement of UPS in periodontitis progression. Increasing evidence indicated that E3s, such as CUL3, Nedd4-2, Synoviolin, FBXL19, PDLIM2, TRIMs and TRAFs, modulate inflammatory responses and bone resorption in periodontitis through multiple classical signalling pathways, including NLRP3, GSDMD, NF-κB, Wnt/β-catenin and Nrf2. Meanwhile, DUBs, including OTUD1, A20, CYLD, UCH-L1 and USPs, also broadly modulate periodontitis progression by regulating signalling pathways such as NF-κB, Wnt/β-catenin, NLRP3, and BMP2. Therefore, the modulation of E3s and DUBs has proven to be an effective therapy against periodontitis. This review provides a comprehensive overview of the regulatory role of ubiquitinating and deubiquitinating enzymes in periodontitis progression and the underlying mechanisms. Finally, we summarise several chemical and genetic methods that regulate UPS enzymes and pave the way for the development of targeted therapies for periodontitis.
Collapse
Affiliation(s)
- Yilin Ma
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Ruiwei Jia
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Shuhong Chen
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Jun Ma
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Lei Yin
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Xingbei Pan
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Yunuo He
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Tong Wu
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Zheyu Zhao
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Lulu Ma
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Shengzhuang Wu
- Institute of Stomatology, School and Hospital of StomatologyHangzhou Medical CollegeZhejiangHangzhouChina
| | - Huining Wang
- Department of Periodontics, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Guang Liang
- Institute of Stomatology, School and Hospital of StomatologyHangzhou Medical CollegeZhejiangHangzhouChina
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
- Department of Prosthodontics, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| | - Xiaoyu Sun
- Institute of Stomatology, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
- Department of Periodontics, School and Hospital of StomatologyWenzhou Medical UniversityWenzhouChina
| |
Collapse
|
12
|
Hernández-Camarero P, Toledo B, Diaz-Ruano AB, González-Titos A, García-Ortega MB, Perán M. What is the Impact of Endothelial-to-Mesenchymal Transition in Solid Tumours: A Qualitative Systematic Review and Quantitative Meta-Analysis. Int J Biol Sci 2025; 21:2155-2178. [PMID: 40083695 PMCID: PMC11900810 DOI: 10.7150/ijbs.107045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/23/2025] [Indexed: 03/16/2025] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) has gained increasing recognition as a crucial mechanism in the progression of solid cancers, influencing tumour heterogeneity, metastasis, and resistance to therapy. However, despite its growing importance, EndMT remains insufficiently studied within the cancer research landscape. In this study, we conduct a systematic review, adhered to the 2020 PRISMA guidelines, of the existing literature on EndMT in solid tumours, examining its functional roles, key biomarkers, underlying mechanisms, experimental models, and potential as a target for therapeutic intervention. Our objective was to identify critical areas where further research is needed. In addition, we performed a meta-analysis to evaluate the variability in the expression of EndMT-related markers and their potential links to patient prognosis. To this aim, literature searches were conducted in major databases including PubMed, Scopus, and Web of Science, covering studies published up to June 2024. The risk of bias of selected articles was evaluated using the OHAT tool, for the in vitro experiments and the SYRCLE tool for studies using animal models. Out of an initial pool of 1,197 articles, 54 studies were selected for data extraction by two independent reviewers. Selected studies were identified according to specific inclusion/exclusion criteria applied through distinct stages like "title and abstract screening", "full text article review" and "article bibliography screening". Our analysis confirms that EndMT is a key contributor to tumour progression and metastasis, but several aspects remain poorly understood, particularly regarding the induction of EndMT in specific cancer types, its role in lymphatic endothelial cells, and its interactions with other stromal elements. We observed substantial heterogeneity in the biomarkers associated with EndMT, as well as variations in the endothelial cell types studied, the functional outcomes, and the molecular mechanisms involved. Our meta-analysis revealed significant variability in the expression of EndMT biomarkers, with notable correlations between changes in the expression of specific genes and patient outcomes, particularly in lung cancer. In conclusion, it is essential for future research to focus on identifying the specific cancer and stromal cell types implicated in EndMT and to standardize endothelial cell models and protocols used for inducing EndMT. Investigating EndMT alongside well-established processes, such as epithelial-to-mesenchymal transition (EMT), and exploring its relationship with cancer-associated fibroblasts (CAFs) may provide valuable insights into its role in tumour biology and its impact on therapy resistance.
Collapse
Affiliation(s)
- Pablo Hernández-Camarero
- Department of Health Sciences, University of Jaén, Campus de las Lagunillas, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, E-18016 Granada, Spain
| | - Belén Toledo
- Department of Health Sciences, University of Jaén, Campus de las Lagunillas, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, E-18016 Granada, Spain
| | - Ana Belén Diaz-Ruano
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, E-18016 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, E-18071 Granada, Spain
| | - Aitor González-Titos
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, E-18016 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, E-18071 Granada, Spain
| | | | - Macarena Perán
- Department of Health Sciences, University of Jaén, Campus de las Lagunillas, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| |
Collapse
|
13
|
Duan Y, Xu Y, Dou Y, Xu D. Helicobacter pylori and gastric cancer: mechanisms and new perspectives. J Hematol Oncol 2025; 18:10. [PMID: 39849657 PMCID: PMC11756206 DOI: 10.1186/s13045-024-01654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Gastric cancer remains a significant global health challenge, with Helicobacter pylori (H. pylori) recognized as a major etiological agent, affecting an estimated 50% of the world's population. There has been a rapidly expanding knowledge of the molecular and pathogenetic mechanisms of H. pylori over the decades. This review summarizes the latest research advances to elucidate the molecular mechanisms underlying the H. pylori infection in gastric carcinogenesis. Our investigation of the molecular mechanisms reveals a complex network involving STAT3, NF-κB, Hippo, and Wnt/β-catenin pathways, which are dysregulated in gastric cancer caused by H. pylori. Furthermore, we highlight the role of H. pylori in inducing oxidative stress, DNA damage, chronic inflammation, and cell apoptosis-key cellular events that pave the way for carcinogenesis. Emerging evidence also suggests the effect of H. pylori on the tumor microenvironment and its possible implications for cancer immunotherapy. This review synthesizes the current knowledge and identifies gaps that warrant further investigation. Despite the progress in our previous knowledge of the development in H. pylori-induced gastric cancer, a comprehensive investigation of H. pylori's role in gastric cancer is crucial for the advancement of prevention and treatment strategies. By elucidating these mechanisms, we aim to provide a more in-depth insights for the study and prevention of H. pylori-related gastric cancer.
Collapse
Affiliation(s)
- Yantao Duan
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yonghu Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Dou
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dazhi Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Fei X, Chen S, Li L, Xu X, Wang H, Ke H, He C, Xie C, Wu X, Liu J, Xie Y, Lu N, Zhu Y, Li N. Helicobacter pylori infection promotes M1 macrophage polarization and gastric inflammation by activation of NLRP3 inflammasome via TNF/TNFR1 axis. Cell Commun Signal 2025; 23:6. [PMID: 39762835 PMCID: PMC11705855 DOI: 10.1186/s12964-024-02017-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Macrophages play a crucial role in chronic gastritis induced by the pathogenic Helicobacter pylori (H. pylori) infection. NLRP3 inflammasome has emerged as an important component of inflammatory processes. However, the molecular mechanism by which H. pylori infection drives NLRP3 inflammasome and macrophages activation remains unclear. METHODS Human gastritis tissues were collected for clinical significance of NLRP3. Infection with H. pylori was performed using in vitro and in vivo models. Bone marrow-derived macrophages (BMDMs) from wild-type (WT), Nlrp3-knockout (KO) and Tnfr1-KO mice were infected with H. pylori. Western blotting, qRT-PCR, immunofluorescence, immunohistochemistry and ELISA were utilized for functional and mechanistic studies. RESULTS Single-cell RNA sequencing (ScRNA-seq) analysis of human gastric tissues, followed by validation, indicated that NLRP3 was primarily expressed in myeloid cells and was significantly increased in H. pylori-positive gastritis compared to H. pylori-negative gastritis. Infection with PMSS1 and NCTC11637 H. pylori strains induced NLRP3 inflammasome activation in vitro (THP1 cells) and in the insulin-gastrin (INS-GAS) transgenic mouse model. Deletion of NLRP3 in BMDMs showed marked inhibition of H. pylori-induced M1 macrophage polarization. Furthermore, NLRP3 inflammasome activation upon TNFα, or H. pylori stimulation, was partially blocked by TNFα/TNFR1 signaling inhibitors. Deletion of TNFR1 in BMDMs significantly impaired NLRP3 inflammasome activation and M1 macrophages induced by H. pylori. CONCLUSION This study revealed that the activation of NLRP3 inflammasome, regulated by the TNF/TNFR1 signaling axis, is a key regulator of H. pylori-induced M1 macrophage activation and gastritis.
Collapse
Affiliation(s)
- Xiao Fei
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Sihai Chen
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Leyan Li
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xinbo Xu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Huan Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Huajing Ke
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Cong He
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Chuan Xie
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xidong Wu
- Department of Drug Safety Evaluation, Jiangxi Testing Center of Medical Instruments, Nanchang, China
| | - Jianping Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yong Xie
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nonghua Lu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Nianshuang Li
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
15
|
Huang M, Ji J, Xu X, Jin D, Wu T, Lin R, Huang Y, Qian J, Tan Z, Jiang F, Hu X, Xu W, Xiao M. Known and unknown: Exosome secretion in tumor microenvironment needs more exploration. Genes Dis 2025; 12:101175. [PMID: 39524543 PMCID: PMC11550746 DOI: 10.1016/j.gendis.2023.101175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/06/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2024] Open
Abstract
Exosomes, extracellular vesicles originating from endosomes, were discovered in the late 1980s and their function in intercellular communication has since garnered considerable interest. Exosomes are lipid bilayer-coated vesicles that range in size from 30 to 150 nm and appear as sacs under the electron microscope. Exosome secretion is crucial for cell-to-cell contact in both normal physiology and the development and spread of tumors. Furthermore, cancer cells can secrete more exosomes than normal cells. Scientists believe that intercellular communication in the complex tissue environment of the human body is an important reason for cancer cell invasion and metastasis. For example, some particles containing regulatory molecules are secreted in the tumor microenvironment, including exosomes. Then the contents of exosomes can be released by donor cells into the environment and interact with recipient cells to promote the migration and invasion of tumor cells. Therefore, in this review, we summarized the biogenesis of exosome, as well as exosome cargo and related roles. More importantly, this review introduces and discusses the factors that have been reported to affect exosome secretion in tumors and highlights the important role of exosomes in tumors.
Collapse
Affiliation(s)
- Mengxiang Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jie Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xuebing Xu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Dandan Jin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Tong Wu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Renjie Lin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Yuxuan Huang
- Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jiawen Qian
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Zhonghua Tan
- Department of Nuclear Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Feng Jiang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaogang Hu
- Department of Respiratory Medicine, Rudong County People's Hospital, Nantong, Jiangsu 226400, China
| | - Weisong Xu
- Department of Gastroenterology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
16
|
Kim JK, Sapkota A, Roh T, Jo EK. The intricate interactions between inflammasomes and bacterial pathogens: Roles, mechanisms, and therapeutic potentials. Pharmacol Ther 2025; 265:108756. [PMID: 39581503 DOI: 10.1016/j.pharmthera.2024.108756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/06/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Inflammasomes are intracellular multiprotein complexes that consist of a sensor, an adaptor, and a caspase enzyme to cleave interleukin (IL)-1β and IL-18 into their mature forms. In addition, caspase-1 and -11 activation results in the cleavage of gasdermin D to form pores, thereby inducing pyroptosis. Activation of the inflammasome and pyroptosis promotes host defense against pathogens, whereas dysregulation of the inflammasome can result in various pathologies. Inflammasomes exhibit versatile microbial signal detection, directly or indirectly, through cellular processes, such as ion fluctuations, reactive oxygen species generation, and the disruption of intracellular organelle function; however, bacteria have adaptive strategies to manipulate the inflammasome by altering microbe-associated molecular patterns, intercepting innate pathways with secreted effectors, and attenuating inflammatory and cell death responses. In this review, we summarize recent advances in the diverse roles of the inflammasome during bacterial infections and discuss how bacteria exploit inflammasome pathways to establish infections or persistence. In addition, we highlight the therapeutic potential of harnessing bacterial immune subversion strategies against acute and chronic bacterial infections. A more comprehensive understanding of the significance of inflammasomes in immunity and their intricate roles in the battle between bacterial pathogens and hosts will lead to the development of innovative strategies to address emerging threats posed by the expansion of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Asmita Sapkota
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Taylor Roh
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea; Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
17
|
Rui Q, Li C, Rui Y, Zhang C, Xia C, Wang Q, Liu Y, Wang P. Human umbilical mesenchymal stem cells ameliorate atrophic gastritis in aging mice by participating in mitochondrial autophagy through Ndufs8 signaling. Stem Cell Res Ther 2024; 15:491. [PMID: 39707499 DOI: 10.1186/s13287-024-04094-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a chronic disease of the gastric mucosa characterized by a reduction or an absolute disappearance of the original gastric glands, possibly replaced by pseudopyloric fibrosis, intestinal metaplasia, or fibrosis. CAG develops progressively into intestinal epithelial metaplasia, dysplasia, and ultimately, gastric cancer. Epidemiological statistics have revealed a positive correlation between the incidence of CAG and age. Mesenchymal stem cells (MSCs) are a type of adult stem cells derived from mesoderm, with strong tissue repair capabilities. Therefore, the restoration of the gastric mucosa may serve as an efficacious strategy to ameliorate CAG and avert gastric cancer. However, the mechanisms by which MSCs inhibit the relentless progression of aging atrophic gastritis remain to be elucidated. This study endeavored to assess a novel approach utilizing MSCs to treat CAG and forestall carcinogenics. METHODS In this study, we selected mice with atrophic gastritis from naturally aging mice and administered human umbilical cord-derived mesenchymal stem cells (hUMSCs) via tail vein injection to evaluate the therapeutic effects of hUMSCs on age-related chronic atrophic gastritis. Initially, we employed methods such as ELISA, immunohistochemical analysis, and TUNEL assays to detect changes in the mice post-hUMSC injection. Proteomic and bioinformatics analyses were conducted to identify differentially expressed proteins, focusing on NADH: ubiquinone oxidoreductase core subunit S8 (Ndufs8). Co-culturing hUMSCs with Ndufs8 knockout gastric mucosal epithelial cells (GMECs), we utilized flow cytometry, Western blotting, real-time quantitative PCR, and immunofluorescence to investigate the mechanisms of action of hUMSCs. RESULTS We observed that hUMSCs are capable of migrating to and repairing damaged gastric mucosa. Initially, hUMSCs significantly enhanced the secretion of gastric proteins PG-1 and G17, while concurrently reducing inflammatory cytokines. Furthermore, hUMSCs mitigated gastric fibrosis and apoptosis in mucosal cells. Proteomic and bioinformatic analyses revealed alterations in the protein network involved in mitochondrial autophagy, with Ndufs8 playing a pivotal role. Upon knocking out Ndufs8 in GMECs, we noted mitochondrial damage and reduced autophagy, leading to an aged phenotype in GMECs. Co-culturing Ndufs8-knockout GMECs with hUMSCs demonstrated that hUMSCs could ameliorate mitochondrial dysfunction and restore the cell cycle in GMECs.
Collapse
Affiliation(s)
- Qiang Rui
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Chuyu Li
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Yiqi Rui
- Department of General Surgery,Geriatric Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Chuanzhuo Zhang
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Cunbing Xia
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Qing Wang
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yuanyuan Liu
- School of Medicine, Southeast University, Nanjing, 210096, China
| | - Peng Wang
- Department of General Surgery,Geriatric Hospital of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
18
|
Xia TS, Xu SY, Lai LY, Jiang YP, Wang NN, Xin HL. Bitter acids from Humulus lupulus L. alleviate D-galactose induced osteoblastic senescence and bone loss via regulating AKT/mTOR-mediated autophagy. J Food Drug Anal 2024; 32:506-519. [PMID: 39752859 PMCID: PMC11698591 DOI: 10.38212/2224-6614.3508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/13/2024] [Indexed: 01/07/2025] Open
Abstract
Bitter acids (BA) are main component of Humulus lupulus L. (hops). They are known for beer brewing and have various biological and pharmacological properties, especially the bone-protective effect confirmed by our previous in vivo study. Here we aimed to elucidate the anti-senior osteoporosis (SOP) effect of BA on osteoblasts and explore its underlying mechanism. In vitro SOP model was established by D-galactose (D-gal) injured osteoblasts, and the bone formation markers and apoptosis level were measured. mCherry-EGFP-LC3 adenovirus infection and autophagic markers including beclin1 and LC3 proteins were detected to investigate the autophagy level in osteoblasts. To further verify whether BA play the bone-protective role through regulating autophagy, the autophagy inhibitor 3-MA was used, and the cell proliferation, ALP activity, bone mineralization, apoptosis rate and SA-β-gal staining areas were measured. Finally, the protein expressions of AKT/mTOR signaling pathway were detected by Western blotting, and AKT agonist SC79 and mTOR agonist MHY1485 were used to further study the mechanism of BA on AKT/mTOR-mediated autophagy. The results showed that BA stimulated osteoblastic differentiation and inhibited apoptosis proteins Bcl-2/Bax in D-gal-treated osteoblasts. BA also increased the expression of autophagic markers beclin1 and LC3-II/LC3-I in D-gal-treated osteoblasts. mCherry-EGFP-LC3 autophagic double fluorescent adenovirus showed BA promoted the generation of autolysosomes and autophagosomes in D-gal-injured osteoblasts, indicating that BA might prevent osteoblastic bone loss through activating autophagy. Autophagy inhibitor 3-MA was used to further verify whether BA played the bone-protective role via regulating autophagy. The results revealed the promotion effects of BA on proliferation, ALP activity, and mineralized nodule formation in D-gal-injured osteoblasts were eliminated after autophagy blocking with 3-MA, and the inhibitory effects of BA on apoptosis rate and SA-β-gal staining areas were also eliminated. Moreover, BA reduced the phosphorylation levels of AKT, mTOR, p70S6K, and 4EBP in AKT/mTOR pathway, and the promotion of BA on the autophagic markers was blocked after the activation of AKT and mTOR by SC79 and MHY1485. In conclusion, it was the first time to demonstrate that BA improved cell activities and bone formation in aging osteoblasts, and revealed the mechanism of BA against SOP in osteoblasts was activating AKT/mTOR-mediated autophagy.
Collapse
Affiliation(s)
- Tian-Shuang Xia
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| | - Sheng-Yan Xu
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| | - Li-Yong Lai
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| | - Yi-Ping Jiang
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| | - Na-Ni Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007,
China
| | - Hai-Liang Xin
- School of Pharmacy, Naval Medical University, Shanghai, 200433,
China
| |
Collapse
|
19
|
Duan F, Li H, Lu B, Wang X, Xu X. Loss of Trim31 Worsens Cardiac Remodeling in a Mouse Model of Heart Failure by Enhancing the Activation of the NLRP3 Inflammasome. Inflammation 2024:10.1007/s10753-024-02217-w. [PMID: 39673012 DOI: 10.1007/s10753-024-02217-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
Tripartite motif-containing protein 31 (Trim31) is known to be involved in various pathological conditions, including heart diseases. Nonetheless, its specific involvement in heart failure (HF) has yet to be determined. In this study, we examined the function and mechanism of Trim31 in HF by using mice with cardiac-specific knockout (cKO) of Trim31. The HF mouse model was induced via the subcutaneous injection of isoproterenol (ISO). We observed a decrease in Trim31 expression in the heart tissues of mice with HF. Compared with wild-type (WT) mice, Trim31 cKO mice presented more severe characteristics of HF, including worsened cardiac dysfunction, hypertrophy, and fibrosis. However, these symptoms in Trim31 cKO mice were significantly reversed when they received an intramyocardial injection of recombinant adeno-associated virus (AAV) expressing Trim31. Excessive activation of the NLRP3 inflammasome, manifested by increased levels of NLRP3, ASC, cleaved Caspase-1, cleaved GSDMD, IL-1β, and IL-18, was observed in Trim31 cKO mice with HF. However, Trim31 overexpression effectively reversed the NLRP3 inflammasome activation in Trim31 cKO mice with HF. Selective inhibition of the NLRP3 inflammasome with the NLRP3 inhibitor MCC950 effectively reversed the worsened cardiac dysfunction, hypertrophy, and fibrosis observed in Trim31 cKO mice with HF. Overall, the findings from this study reveal a crucial role of Trim31 in HF. Trim31 deficiency may contribute to the progression of HF by promoting cardiac hypertrophy, fibrosis, and inflammation by facilitating the activation of the NLRP3 inflammasome. Therefore, Trim31 may hold significant potential as a therapeutic target for the treatment of HF.
Collapse
Affiliation(s)
- Fengqi Duan
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518106, Guangdong, China
| | - Huangjing Li
- Department of Pathophysiology, School of Medicine, Sun Yat-Sen University, Shenzhen, 518106, Guangdong, China
| | - Bo Lu
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518106, Guangdong, China
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518106, Guangdong, China
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518106, Guangdong, China.
| |
Collapse
|
20
|
Fan H, Xu P, Zou B, Wang H, Li C, Huang J. Isoquercitrin Inhibits Lung Cancer Cell Growth Through Triggering Pyroptosis and Ferroptosis. Nutr Cancer 2024; 77:299-310. [PMID: 39427296 DOI: 10.1080/01635581.2024.2416246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Isoquercitrin possesses anti-tumor activity in several types of cancers, however, its effects and underlying mechanisms on lung cancer have not been reported. Human lung cancer cell lines as well as normal lung epithelial BEAS-2B cells were treated with isoquercitrin. The influences of isoquercitrin in vitro were evaluated by determining cell viability, apoptosis, pyroptosis, and ferroptosis. Additionally, A549 tumor-bearing mice were generated to explore the anti-cancer effect of isoquercitrin in vivo. We found that isoquercitrin dose-dependently reduced lung cancer cells' viability, with no toxicity against BEAS-2B cells. Isoquercitrin at 40 μM and 80 μM was used in vitro. Isoquercitrin increased apoptosis, elevated NLRP3 inflammasome activation-mediated pyroptosis, and promoted ferroptosis in lung cancer cells. NLRP3 knockdown and caspase-1 selective inhibitor VX-765 attenuated isoquercitrin-induced pyroptosis and ferroptosis, but not apoptosis. Furthermore, isoquercitrin accelerated ROS generation, while ROS inhibitor N-acetylcysteine abrogated isoquercitrin-induced apoptosis, NLRP3 related-pyroptosis and ferroptosis. In vivo, isoquercitrin (1 mg/kg and 5 mg/kg) inhibited tumor growth, increased apoptosis, NLRP3-related pyroptosis, ferroptosis and ROS generation in tumors. Taken together, isoquercitrin inhibits lung cancer growth by triggering ROS/NLRP3-mediated pyroptosis and ferroptosis, with ROS also directly inducing apoptosis. This suggests that isoquercitrin might be a potential therapeutic agent for lung cancer.
Collapse
Affiliation(s)
- Haiyin Fan
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi Province, China
| | - Pengfei Xu
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi Province, China
| | - Bin Zou
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi Province, China
| | - Huanyuan Wang
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi Province, China
| | - Chao Li
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi Province, China
| | - Jian Huang
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi Province, China
| |
Collapse
|
21
|
Mottaghi M, Eidi A, Heidari F, Movahhed TK, Moslehi A. SIRT1/NOX1 pathway mediated ameliorative effects of rosmarinic acid in folic acid-induced renal injury. Res Pharm Sci 2024; 19:622-633. [PMID: 39691298 PMCID: PMC11648342 DOI: 10.4103/rps.rps_213_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 04/24/2024] [Accepted: 08/18/2024] [Indexed: 12/19/2024] Open
Abstract
Background and purpose Renal injury is a serious disorder that can be caused by some diseases or agents. Rosmarinic acid (RA) is a natural and safe compound with powerful antioxidant and anti-inflammatory properties. In this study, the ameliorative effects of RA were assayed in folic acid (FA)-induced renal injury by involving the SIRT1/NOX1 pathway. Experimental approach Thirty-six male C57/BL6 mice were divided into 6 groups (n = 6) including control, vehicle, FA, RA, FA + RA 50, and FA + RA 100. After 10 days, blood urea nitrogen (BUN), creatinine, and oxidative stress were measured. The expression of SIRT1 and NOX1 proteins was evaluated by western blot. Also, histopathological alterations were assayed by H&E and PAS staining methods. Findings/Results BUN and creatinine were significantly higher in the FA group compared to the control group; however, their levels decreased after RA treatment in both doses. A significant decrease was observed in swelling, necrosis, and desquamation of tubular epithelial cells in the FA + RA 50 and FA + RA 100 groups compared to the FA group. RA in the animals receiving FA increased SIRT1 expression and the levels of GSH and SOD compared to the FA group. RA in the animals receiving FA showed a significant decrease in NOX1 expression and MDA level compared to the FA group. Conclusion and implications The findings declared that the administration of RA has positive effects against renal damage induced by FA. The effect might result from involvement in the SIRT1/NOX1 pathway and thereby attenuation of oxidative stress.
Collapse
Affiliation(s)
- Maryam Mottaghi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, I.R. Iran
| | - Akram Eidi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, I.R. Iran
| | - Fatemeh Heidari
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, I.R. Iran
| | | | - Azam Moslehi
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, I.R. Iran
- Department of Physiolgy, Faculty of Medicine, Qom University of Medical Sciences, Qom, I.R. Iran
| |
Collapse
|
22
|
Xue J, Jiang C, Chen X, Wang L. Trim31 deficiency exacerbates airway inflammation in asthma by enhancing the activation of the NLRP3 inflammasome. Int Immunopharmacol 2024; 138:112591. [PMID: 38981220 DOI: 10.1016/j.intimp.2024.112591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/23/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024]
Abstract
Tripartite motif (Trim) 31 is important for numerous inflammatory diseases. However, whether Trim31 regulates airway inflammation in asthma remains undetermined. The present work explored the role of Trim31 in airway inflammation in asthmatic mice established by ovalbumin (OVA) stimulation. Trim31 expression was markedly downregulated in the lungs of asthmatic mice. Compared with wild-type (WT) mice, Trim31-/- mice showed more severe pathological changes accompanied by increased inflammatory cell infiltration after OVA induction. House dust mite (HDM) stimulation evoked airway epithelial cell injury and inflammation, which were exacerbated by Trim31 silencing or attenuated by Trim31 overexpression. Further examination revealed that Trim31 deficiency exacerbated the activation of the NLRP3 inflammasome in OVA-induced asthmatic mice and HDM-stimulated airway epithelial cells. The inhibition of NLRP3 markedly diminished the Trim31 silencing-mediated enhancement of HDM-induced injury and inflammation in airway epithelial cells. In conclusion, this work demonstrates that Trim31 acts as a crucial mediator of airway inflammation in asthma. Trim31 deficiency may contribute to the progression of asthma by increasing NLRP3 inflammasome activation, suggesting that Trim31 is a potential therapeutic target for asthma.
Collapse
Affiliation(s)
- Jing Xue
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China.
| | - Chunyan Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Xue Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| | - Li Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, China
| |
Collapse
|
23
|
Lin CJ, Tian GA, Zhao WY, Tian Y, Liu YR, Gu DN, Tian L. NLRP1 inhibits lung adenocarcinoma growth through mediating mitochondrial dysregulation in an inflammasome-independent manner. Braz J Med Biol Res 2024; 57:e13885. [PMID: 39258674 PMCID: PMC11379352 DOI: 10.1590/1414-431x2024e13885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/21/2024] [Indexed: 09/12/2024] Open
Abstract
NLRP1, the first identified inflammasome-forming sensor, is thought to be involved in cancer, yet its definite function in lung adenocarcinoma (LUAD) remains unclear. Herein, we explored the expression and function of NLRP1 in LUAD. Decreased NLRP1 expression was identified in LUAD, which was associated with a poor prognosis. Overexpression of NLRP1 inhibited tumor growth in vitro and in vivo. Mechanically, this effect was observed regardless of inflammasome activation. Further studies revealed that overexpression of NLRP1 downregulated the phosphorylation of DRP1 and promoted mitochondrial fusion, which was mediated by inhibition of NF-κB activity. NF-κB agonist could neutralize the effect of NLRP1 on mitochondrial dynamics. In addition, LUAD sensitivity to cisplatin was enhanced by decreased mitochondrial fission resulting from up-regulated NLRP1. In conclusion, our findings demonstrated an unexpected role of NLRP1 in LUAD by modulating mitochondrial activities, which provides strong evidence for its potential in LUAD treatment.
Collapse
Affiliation(s)
- Chen-jing Lin
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang-ang Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-ya Zhao
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-ru Liu
- Department of Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dian-na Gu
- Department of Chemotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ling Tian
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Liu J, Xiao Y, Xu Q, Xu Y, Guo M, Hu Y, Wang Y, Wang Y. Britannilactone 1-O-acetate induced ubiquitination of NLRP3 inflammasome through TRIM31 as a protective mechanism against reflux esophagitis-induced esophageal injury. Chin Med 2024; 19:118. [PMID: 39215331 PMCID: PMC11363507 DOI: 10.1186/s13020-024-00986-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Reflux esophagitis (RE) is a disease in which inflammation of the esophageal mucosa owing to the reflux of gastric contents into the esophagus results in cytokine damage. Britannilactone 1-O-acetate (Brt) has anti-inflammatory effects, significantly inhibiting the activation of the NLRP3 inflammasome, leading to a decrease in inflammatory factors including IL-1 β, IL-6, and TNF-α. However, the mechanism underlying its protective effect against RE-induced esophageal injury remains unclear. In the present study, we investigated the protective mechanism of TRIM31 against NLRP3 ubiquitination-induced RE both in vivo and in vitro. METHODS A model of RE was established in vivo in rats by the method of "4.2 mm pyloric clamp + 2/3 fundoplication". In vitro, the mod was constructed by using HET-1A (esophageal epithelial cells) and exposing the cells to acid, bile salts, and acidic bile salts. The 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay was used to screen the concentration of administered drugs, and the viability of HET-1A cells in each group. HE staining was used to assess the degree of pathological damage in esophageal tissues. Toluidine blue staining was used to detect whether the protective function of the esophageal epithelial barrier was damaged and restored. The enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of IL-1 β, IL-6, and TNF-α factors in serum. Immunohistochemistry (IHC) was used to detect the expression level of NLRP3 in esophageal tissues. The molecular docking and Co-immunoprecipitation assay (Co-IP assay) were used to detect the TRIM31 interacts with NLRP3. Western blotting detected the Claudin-4, Claudin-5, The G-protein-coupled receptor calcium-sensitive receptor (CaSR), NLRP3, TRIM31, ASC, C-Caspase1, and Caspase1 protein expression levels. RESULTS Brt could alleviate RE inflammatory responses by modulating serum levels of IL-1 β, IL-6, and TNF-α. It also activated the expression of NLRP3, ASC, Caspase 1, and C-Caspase-1 in HET-1A cells. Brt also attenuated TRIM31/NLRP3-induced pathological injury in rats with RE through a molecular mechanism consistent with the in vitro results. CONCLUSIONS Brt promotes the ubiquitination of NLRP3 through TRIM31 and attenuates esophageal epithelial damage induced by RE caused by acidic bile salt exposure. This study provides valuable insights into the mechanism of action of Brt in the treatment of RE and highlights its promising application in the prevention of NLRP3 inflammatory vesicle-associated inflammatory pathological injury.
Collapse
Affiliation(s)
- Ju Liu
- Office of Science and Technology Administration, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Yang Xiao
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qianfei Xu
- Department of Spleen, Stomach and Hepatobiliary, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Yunyan Xu
- Preventive Treatment Department, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Manman Guo
- Pharmaceutical Department, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Yun Hu
- Department of Spleen, Stomach and Hepatobiliary, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Yan Wang
- Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yi Wang
- Pharmaceutical Department, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China.
| |
Collapse
|
25
|
Zhang X, Zhang B, Zhang Y, Ding Y, Zhang Z, Liu Q, Yang Z, Wang L, Gao J. Copper-Induced Supramolecular Peptide Assemblies for Multi-Pathway Cell Death and Tumor Inhibition. Angew Chem Int Ed Engl 2024; 63:e202406602. [PMID: 38837577 DOI: 10.1002/anie.202406602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Although self-assembly has emerged as an effective tool for fabricating biomaterials, achieving precise control over the morphologies and functionalities of the resultant assemblies remains an ongoing challenge. Inspired by the copper peptide naturally present in human plasma, in this study, we designed a synthetic precursor, FcGH. FcGH can self-assemble via two distinct pathways: spontaneous and Cu2+-induced. These two assembly pathways enabled the formation of assemblies with tunable morphologies by adjusting the amount of added Cu2+. We found that the nanoparticles formed by Cu2+-induced self-assembly exhibited a significantly higher cellular uptake efficiency than the wormlike fibers formed spontaneously. Moreover, this Cu2+-induced assembly process occurred spontaneously at a 1 : 1 molar ratio of Cu2+ to FcGH, avoiding the excessive use of Cu2+ and a tedious preparation procedure. By co-assembling with 10-hydroxycamptothecin (HCPT)-conjugated FcGH, Cu2+-induced supramolecular nanodrugs elicited multiple cell death modalities in cancer cells with elevated immunogenicity, enhancing the therapeutic effect compared to free HCPT. This study highlights Cu2+-induced self-assembly as an efficient tool for directing the assembly of nanodrugs and for synergistic tumor therapy.
Collapse
Affiliation(s)
- Xiangyang Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Buyue Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Ying Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Yinghao Ding
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Zhenghao Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University Xuzhou, Jiangsu, 221002, China
| | - Ling Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai International Advanced Research Institute (SHENZHEN⋅FUTIAN), Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Tianjin, 300071, China
| |
Collapse
|
26
|
Shang A, Shao S, Zhao L, Liu B. Far-Red Fluorescent Proteins: Tools for Advancing In Vivo Imaging. BIOSENSORS 2024; 14:359. [PMID: 39194588 DOI: 10.3390/bios14080359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024]
Abstract
Far-red fluorescent proteins (FPs) have emerged as indispensable tools in in vivo imaging, playing a pivotal role in elucidating fundamental mechanisms and addressing application issues in biotechnology and biomedical fields. Their ability for deep penetration, coupled with reduced light scattering and absorption, robust resistance to autofluorescence, and diminished phototoxicity, has positioned far-red biosensors at the forefront of non-invasive visualization techniques for observing intracellular activities and intercellular behaviors. In this review, far-red FPs and their applications in living systems are mainly discussed. Firstly, various far-red FPs, characterized by emission peaks spanning from 600 nm to 650 nm, are introduced. This is followed by a detailed presentation of the fundamental principles enabling far-red biosensors to detect biomolecules and environmental changes. Furthermore, the review accentuates the superiority of far-red FPs in multi-color imaging. In addition, significant emphasis is placed on the value of far-red FPs in improving imaging resolution, highlighting their great contribution to the advancement of in vivo imaging.
Collapse
Affiliation(s)
- Angyang Shang
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
- Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
| | - Shuai Shao
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
- Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
| | - Luming Zhao
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
- Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
| | - Bo Liu
- Cancer Hospital of Dalian University of Technology, Shenyang 110042, China
- Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
27
|
Güven Gülhan Ü, Nikerel E, Çakır T, Erdoğan Sevilgen F, Durmuş S. Species-level identification of enterotype-specific microbial markers for colorectal cancer and adenoma. Mol Omics 2024; 20:397-416. [PMID: 38780313 DOI: 10.1039/d4mo00016a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Enterotypes have been shown to be an important factor for population stratification based on gut microbiota composition, leading to a better understanding of human health and disease states. Classifications based on compositional patterns will have implications for personalized microbiota-based solutions. There have been limited enterotype based studies on colorectal adenoma and cancer. Here, an enterotype-based meta-analysis of fecal shotgun metagenomic studies was performed, including 1579 samples of healthy controls (CTR), colorectal adenoma (ADN) and colorectal cancer (CRC) in total. Gut microbiota of healthy people were clustered into three enterotypes (Ruminococcus-, Bacteroides- and Prevotella-dominated enterotypes). Reference-based enterotype assignments were performed for CRC and ADN samples, using the supervised machine learning algorithm, K-nearest neighbors. Differential abundance analyses and random forest classification were conducted on each enterotype between healthy controls and CRC-ADN groups, revealing novel enterotype-specific microbial markers for non-invasive CRC screening strategies. Furthermore, we identified microbial species unique to each enterotype that play a role in the production of secondary bile acids and short-chain fatty acids, unveiling the correlation between cancer-associated gut microbes and dietary patterns. The enterotype-based approach in this study is promising in elucidating the mechanisms of differential gut microbiome profiles, thereby improving the efficacy of personalized microbiota-based solutions.
Collapse
Affiliation(s)
- Ünzile Güven Gülhan
- Department of Bioengineering, Gebze Technical University, Gebze, TR 41400, Turkey.
| | - Emrah Nikerel
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, TR 34755, Turkey
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Gebze, TR 41400, Turkey.
- PhiTech Bioinformatics, Gebze, TR 41470, Turkey
| | - Fatih Erdoğan Sevilgen
- The Institute for Data Science & Artificial Intelligence, Boğaziçi University, Istanbul, TR 34342, Turkey
- PhiTech Bioinformatics, Gebze, TR 41470, Turkey
| | - Saliha Durmuş
- Department of Bioengineering, Gebze Technical University, Gebze, TR 41400, Turkey.
- PhiTech Bioinformatics, Gebze, TR 41470, Turkey
| |
Collapse
|
28
|
Zhou C, Cai Z, Guo J, Li C, Qin C, Yan J, Yang D. Injective hydrogel loaded with liposomes-encapsulated MY-1 promotes wound healing and increases tensile strength by accelerating fibroblast migration via the PI3K/AKT-Rac1 signaling pathway. J Nanobiotechnology 2024; 22:396. [PMID: 38965546 PMCID: PMC11225333 DOI: 10.1186/s12951-024-02666-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
Failed skin wound healing, through delayed wound healing or wound dehiscence, is a global public health issue that imposes significant burdens on individuals and society. Although the application of growth factor is an effective method to improve the pace and quality of wound healing, the clinically approved factors are limited. Parathyroid hormone (PTH) demonstrates promising results in wound healing by promoting collagen deposition and cell migration, but its application is limited by potentially inhibitory effects when administered continuously and locally. Through partially replacing and repeating the amino acid domains of PTH(1-34), we previously designed a novel PTH analog, PTH(3-34)(29-34) or MY-1, and found that it avoided the inhibitory effects of PTH while retaining its positive functions. To evaluate its role in wound healing, MY-1 was encapsulated in liposomes and incorporated into the methacryloyl gelatin (GelMA) hydrogel, through which an injectable nanocomposite hydrogel (GelMA-MY@Lipo, or GML) was developed. In vitro studies revealed that the GML had similar properties in terms of the appearance, microstructure, functional groups, swelling, and degradation capacities as the GelMA hydrogel. In vitro drug release testing showed a relatively more sustainable release of MY-1, which was still detectable in vivo 9 days post-application. When the GML was topically applied to the wound areas of rat models, wound closure as well as tensile strength were improved. Further studies showed that the effects of GML on wound repair and tensile strength were closely related to the promotion of fibroblast migration to the wound area through the controlled release of MY-1. Mechanically, MY-1 enhanced fibroblast migration by activating PI3K/AKT signaling and its downstream molecule, Rac1, by which it increased fibroblast aggregation in the early stage and resulting in denser collagen deposition at a later time. Overall, these findings demonstrated that the nanocomposite hydrogel system promoted skin wound healing and increased tensile strength, thus offering new potential in the treatment of wound healing.
Collapse
Affiliation(s)
- Chunhao Zhou
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Zhihai Cai
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Jialiang Guo
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Chengfu Li
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China
| | - Chenghe Qin
- Department of Orthopedics - Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Juanwen Yan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Dehong Yang
- Department of Orthopedics - Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Avenue, Guangzhou, 510515, China.
| |
Collapse
|
29
|
Deng NH, Tian Z, Zou YJ, Quan SB. E3 ubiquitin ligase TRIM31: A potential therapeutic target. Biomed Pharmacother 2024; 176:116846. [PMID: 38850648 DOI: 10.1016/j.biopha.2024.116846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024] Open
Abstract
Ubiquitination is a key mechanism for post-translational protein modification, affecting protein localization, metabolism, degradation and various cellular physiological processes. Dysregulation of ubiquitination is associated with the pathogenesis of various diseases, such as tumors and cardiovascular diseases, making it a primary area of interest in biochemical research and drug development endeavors. E3 ubiquitin ligases play a pivotal role in modulating the ubiquitination of substrate proteins through their unique recognition functions. TRIM31, a member of the TRIM family of E3 ubiquitin ligases, is aberrantly expressed in different pathophysiological conditions. The biological function of TRIM31 is associated with the occurrence and development of diverse diseases. TRIM31 has been demonstrated to inhibit inflammation by promoting ubiquitin-proteasome-mediated degradation of the sensing protein NLRP3 in the inflammasome. TRIM31 mediates ubiquitination of MAVS, inducing the formation of prion-like aggregates, and triggering innate antiviral immune responses. TRIM31 is also implicated in tumor pathophysiology through its ability to promote ubiquitination of the tumor suppressor protein p53. These findings indicate that TRIM31 is a potential therapeutic target, and subsequent in-depth research of TRIM31 is anticipated to provide information on its clinical application in therapy.
Collapse
Affiliation(s)
- Nian-Hua Deng
- The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong 523326, PR China
| | - Zhen Tian
- The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong 523326, PR China
| | - Ying-Jiao Zou
- Medical Technology Center, Shilong Town Community Health Service Center, Dongguan, Guangdong 523326, PR China
| | - Shou-Bo Quan
- The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong 523326, PR China.
| |
Collapse
|
30
|
Nenasheva VV, Stepanenko EA, Tarantul VZ. Multi-Directional Mechanisms of Participation of the TRIM Gene Family in Response of Innate Immune System to Bacterial Infections. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1283-1299. [PMID: 39218025 DOI: 10.1134/s0006297924070101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/30/2024] [Accepted: 06/08/2024] [Indexed: 09/04/2024]
Abstract
The multigene TRIM family is an important component of the innate immune system. For a long time, the main function of the genes belonging to this family was believed to be an antiviral defense of the host organism. The issue of their participation in the immune system response to bacterial invasion has been less studied. This review is the first comprehensive analysis of the mechanisms of functioning of the TRIM family genes in response to bacterial infections, which expands our knowledge about the role of TRIM in the innate immune system. When infected with different types of bacteria, individual TRIM proteins regulate inflammatory, interferon, and other responses of the immune system in the cells, and also affect autophagy and apoptosis. Functioning of TRIM proteins in response to bacterial infection, as well as viral infection, often includes ubiquitination and various protein-protein interactions with both bacterial proteins and host cell proteins. At the same time, some TRIM proteins, on the contrary, contribute to the infection development. Different members of the TRIM family possess similar mechanisms of response to viral and bacterial infection, and the final impact of these proteins could vary significantly. New data on the effect of TRIM proteins on bacterial infections make an important contribution to a more detailed understanding of the innate immune system functioning in animals and humans when interacting with pathogens. This data could also be used for the search of new targets for antibacterial defense.
Collapse
|
31
|
Ju M, Deng T, Jia X, Gong M, Li Y, Liu F, Yin Y. The causal relationship between anti-diabetic drugs and gastrointestinal disorders: a drug-targeted mendelian randomization study. Diabetol Metab Syndr 2024; 16:141. [PMID: 38918852 PMCID: PMC11201305 DOI: 10.1186/s13098-024-01359-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND The incidence of diabetic gastrointestinal diseases is increasing year by year. This study aimed to investigate the causal relationship between antidiabetic medications and gastrointestinal disorders, with the goal of reducing the incidence of diabetes-related gastrointestinal diseases and exploring the potential repurposing of antidiabetic drugs. METHODS We employed a two-sample Mendelian randomization (TSMR) design to investigate the causal association between antidiabetic medications and gastrointestinal disorders, including gastroesophageal reflux disease (GERD), gastric ulcer (GU), chronic gastritis, acute gastritis, Helicobacter pylori infection, gastric cancer (GC), functional dyspepsia (FD), irritable bowel syndrome (IBS), ulcerative colitis (UC), Crohn's disease (CD), diverticulosis, and colorectal cancer (CRC). To identify potential inhibitors of antidiabetic drug targets, we collected single-nucleotide polymorphisms (SNPs) associated with metformin, GLP-1 receptor agonists, SGLT2 inhibitors, DPP-4 inhibitors, insulin, and its analogs, thiazolidinediones, sulfonylureas, and alpha-glucosidase inhibitors from published genome-wide association study statistics. We then conducted a drug-target Mendelian randomization (MR) analysis using inverse variance weighting (IVW) as the primary analytical method to assess the impact of these inhibitors on gastrointestinal disorders. Additionally, diabetes was selected as a positive control. RESULTS Sulfonylureas were found to significantly reduce the risk of CD (IVW: OR [95% CI] = 0.986 [0.978, 0.995], p = 1.99 × 10- 3), GERD (IVW: OR [95% CI] = 0.649 [0.452, 0.932], p = 1.90 × 10- 2), and chronic gastritis (IVW: OR [95% CI] = 0.991 [0.982, 0.999], p = 4.50 × 10- 2). However, they were associated with an increased risk of GU development (IVW: OR [95%CI] = 2 0.761 [1.259, 6.057], p = 1 0.12 × 10- 2). CONCLUSIONS The results indicated that sulfonylureas had a positive effect on the prevention of CD, GERD, and chronic gastritis but a negative effect on the development of gastric ulcers. However, our research found no causal evidence for the impact of metformin, GLP-1 agonists, SGLT2 inhibitors, DPP 4 inhibitors, insulin and its analogs, thiazolidinediones, or alpha-glucosidase inhibitors on gastrointestinal diseases.
Collapse
Affiliation(s)
- Mingyan Ju
- College of Acupuncture and moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tingting Deng
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuemin Jia
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Menglin Gong
- College of Acupuncture and moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuying Li
- College of Acupuncture and moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fanjie Liu
- Bone Biomechanics Engineering Laboratory of Shandong Province, Shandong Medicinal Biotechnology Center (School of Biomedical Sciences), Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Ying Yin
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
32
|
Yuan C, Yu C, Sun Q, Xiong M, Ren B, Zhong M, Peng Q, Zeng M, Meng P, Li L, Song H. Atractylenolide I Alleviates Indomethacin-Induced Gastric Ulcers in Rats by Inhibiting NLRP3 Inflammasome Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14165-14176. [PMID: 38872428 DOI: 10.1021/acs.jafc.3c08188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Atractylodes macrocephala Koidz, a traditional Chinese medicine, contains atractylenolide I (ATR-I), which has potential anticancer, anti-inflammatory, and immune-modulating properties. This study evaluated the therapeutic potential of ATR-I for indomethacin (IND)-induced gastric mucosal lesions and its underlying mechanisms. Noticeable improvements were observed in the histological morphology and ultrastructures of the rat gastric mucosa after ATR-I treatment. There was improved blood flow, a significant decrease in the expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), IL-1β, and IL-18, and a marked increase in prostaglandin E2 (PGE2) expression in ATR-I-treated rats. Furthermore, there was a significant decrease in the mRNA and protein expression levels of NOD-like receptor thermal protein domain associated protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), cysteinyl aspartate specific proteinase-1 (caspase-1), and nuclear factor-κB (NF-κB) in rats treated with ATR-I. The results show that ATR-I inhibits the NLRP3 inflammasome signaling pathway and effectively alleviates local inflammation, thereby improving the therapeutic outcomes against IND-induced gastric ulcers in rats.
Collapse
Affiliation(s)
- Chengzhi Yuan
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Chang Yu
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Qifang Sun
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Meng Xiong
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Baoping Ren
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Meiqi Zhong
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Qinghua Peng
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Meiyan Zeng
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Pan Meng
- School of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Liang Li
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Houpan Song
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
33
|
Xing F, Liu N, Wang C, Wang XD. Caffeic acid phenethyl ester promotes oxaliplatin sensitization in colon cancer by inhibiting autophagy. Sci Rep 2024; 14:14624. [PMID: 38918541 PMCID: PMC11199620 DOI: 10.1038/s41598-024-65409-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Colon cancer ranks as the third most prevalent form of cancer globally, with chemotherapy remaining the primary treatment modality. To mitigate drug resistance and minimize adverse effects associated with chemotherapy, selection of appropriate adjuvants assumes paramount importance. Caffeic acid phenethyl ester (CAPE), a naturally occurring compound derived from propolis, exhibits a diverse array of biological activities. We observed that the addition of CAPE significantly augmented the drug sensitivity of colon cancer cells to oxaliplatin. In SW480 and HCT116 cells, oxaliplatin combined with 10 µM CAPE reduced the IC50 of oxaliplatin from 14.24 ± 1.03 and 84.16 ± 3.02 µM to 2.11 ± 0.15 and 3.92 ± 0.17 µM, respectively. We then used proteomics to detect differentially expressed proteins in CAPE-treated SW480 cells and found that the main proteins showing changes in expression after CAPE treatment were p62 (SQSTM1) and LC3B (MAP1LC3B). Gene ontology analysis revealed that CAPE exerted antitumor and chemotherapy-sensitization effects through the autophagy pathway. We subsequently verified the differentially expressed proteins using immunoblotting. Simultaneously, the autophagy inhibitor bafilomycin A1 and the mCherry-EGFP-LC3 reporter gene were used as controls to detect the effect of CAPE on autophagy levels. Collectively, the results indicate that CAPE may exert antitumor and chemotherapy-sensitizing effects by inhibiting autophagy, offering novel insights for the development of potential chemosensitizing agents.
Collapse
Affiliation(s)
- Fei Xing
- Department of Gastrointestinal Nutrition Surgery, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Ning Liu
- Academic Center, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Can Wang
- Department of Gastrointestinal Nutrition Surgery, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Xu-Dong Wang
- Department of Gastrointestinal Nutrition Surgery, The Second Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
34
|
Iqbal I, Saqib F, Mubarak Z, Latif MF, Wahid M, Nasir B, Shahzad H, Sharifi-Rad J, Mubarak MS. Alzheimer's disease and drug delivery across the blood-brain barrier: approaches and challenges. Eur J Med Res 2024; 29:313. [PMID: 38849950 PMCID: PMC11161981 DOI: 10.1186/s40001-024-01915-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
Alzheimer's disease (AD) is a diverse disease with a complex pathophysiology. The presence of extracellular β-amyloid deposition as neuritic plaques and intracellular accumulation of hyper-phosphorylated tau as neurofibrillary tangles remain the core neuropathologic criteria for diagnosing Alzheimer's disease. Nonetheless, several recent basic discoveries have revealed significant pathogenic roles for other essential cellular and molecular processes. Previously, there were not so many disease-modifying medications (DMT) available as drug distribution through the blood-brain barrier (BBB) is difficult due to its nature, especially drugs of polypeptides nature and proteins. Recently FDA has approved lecanemab as DMT for its proven efficacy. It is also complicated to deliver drugs for diseases like epilepsy or any brain tumor due to the limitations of the BBB. After the advancements in the drug delivery system, different techniques are used to transport the medication across the BBB. Other methods are used, like enhancement of brain blood vessel fluidity by liposomes, infusion of hyperosmotic solutions, and local intracerebral implants, but these are invasive approaches. Non-invasive approaches include the formulation of nanoparticles and their coating with polymers. This review article emphasizes all the above-mentioned techniques, procedures, and challenges to transporting medicines across the BBB. It summarizes the most recent literature dealing with drug delivery across the BBB.
Collapse
Affiliation(s)
- Iram Iqbal
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
- Primary and Secondary Healthcare Department, Govt of the Punjab, Lahore, Pakistan
| | - Fatima Saqib
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| | - Zobia Mubarak
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
- Primary and Secondary Healthcare Department, Govt of the Punjab, Lahore, Pakistan
| | - Muhammad Farhaj Latif
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Muqeet Wahid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Bushra Nasir
- Department of Pharmaceutics, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Hamna Shahzad
- Department of Biochemistry, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Javad Sharifi-Rad
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| | - Mohammad S Mubarak
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
35
|
El Menshawe SF, Shalaby K, Elkomy MH, Aboud HM, Ahmed YM, Abdelmeged AA, Elkarmalawy M, Abou Alazayem MA, El Sisi AM. Repurposing celecoxib for colorectal cancer targeting via pH-triggered ultra-elastic nanovesicles: Pronounced efficacy through up-regulation of Wnt/β-catenin pathway in DMH-induced tumorigenesis. Int J Pharm X 2024; 7:100225. [PMID: 38230407 PMCID: PMC10788539 DOI: 10.1016/j.ijpx.2023.100225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 01/18/2024] Open
Abstract
Celecoxib (CLX), a selective inhibitor for cyclooxygenase 2 (COX-2), has manifested potential activity against diverse types of cancer. However, low bioavailability and cardiovascular side effects remain the major challenges that limit its exploitation. In this work, we developed ultra-elastic nanovesicles (UENVs) with pH-triggered surface charge reversal traits that could efficiently deliver CLX to colorectal segments for snowballed tumor targeting. CLX-UENVs were fabricated via a thin-film hydration approach. The impact of formulation factors (Span 80, Tween 80, and sonication time) on the nanovesicular features was evaluated using Box-Behnken design, and the optimal formulation was computed. The optimum formulation was positively coated with polyethyleneimine (CLX-PEI-UENVs) and then coated with Eudragit S100 (CLX-ES-PEI-UENVs). The activity of the optimized nano-cargo was explored in 1,2-dimethylhydrazine-induced colorectal cancer in Wistar rats. Levels of COX-2, Wnt-2 and β-catenin were assessed in rats' colon. The diameter of the optimized CLX-ES-PEI-UENVs formulation was 253.62 nm, with a zeta potential of -23.24 mV, 85.64% entrapment, and 87.20% cumulative release (24 h). ES coating hindered the rapid release of CLX under acidic milieu (stomach and early small intestine) and showed extended release in the colon section. In colonic environments, the ES coating layer was removed due to high pH, and the charge on the nanovesicular corona was shifted from negative to positive. Besides, a pharmacokinetics study revealed that CLX-ES-PEI-UENVs had superior oral bioavailability by 2.13-fold compared with CLX suspension. Collectively, these findings implied that CLX-ES-PEI-UENVs could be a promising colorectal-targeted nanoplatform for effective tumor management through up-regulation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Shahira F. El Menshawe
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Khaled Shalaby
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Mohammed H. Elkomy
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Heba M. Aboud
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Yasmin M. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | | | - Marwa Elkarmalawy
- Department of Pharmaceutics and Drug Manufacturing, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | | | - Amani M. El Sisi
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
36
|
Shen J, Gong L, Sun Y, Lin J, Hu W, Wei J, Miao X, Gao T, Suo J, Xu J, Chai Y, Bao B, Qian Y, Zheng X. Semaphorin3C identified as mediator of neuroinflammation and microglia polarization after spinal cord injury. iScience 2024; 27:109649. [PMID: 38638567 PMCID: PMC11025009 DOI: 10.1016/j.isci.2024.109649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/01/2024] [Accepted: 03/27/2024] [Indexed: 04/20/2024] Open
Abstract
Excessive neuroinflammation after spinal cord injury (SCI) is a major hurdle during nerve repair. Although proinflammatory macrophage/microglia-mediated neuroinflammation plays important roles, the underlying mechanism that triggers neuroinflammation and aggravating factors remain unclear. The present study identified a proinflammatory role of semaphorin3C (SEMA3C) in immunoregulation after SCI. SEMA3C expression level peaked 7 days post-injury (dpi) and decreased by 14 dpi. In vivo and in vitro studies revealed that macrophages/microglia expressed SEMA3C in the local microenvironment, which induced neuroinflammation and conversion of proinflammatory macrophage/microglia. Mechanistic experiments revealed that RAGE/NF-κB was downstream target of SEMA3C. Inhibiting SEMA3C-mediated RAGE signaling considerably suppressed proinflammatory cytokine production, reversed polarization of macrophages/microglia shortly after SCI. In addition, inhibition of SEMA3C-mediated RAGE signaling suggested that the SEMA3C/RAGE axis is a feasible target to preserve axons from neuroinflammation. Taken together, our study provides the first experimental evidence of an immunoregulatory role for SEMA3C in SCI via an autocrine mechanism.
Collapse
Affiliation(s)
- Junjie Shen
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Liangzhi Gong
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Yi Sun
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Junqing Lin
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Wencheng Hu
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Jiabao Wei
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Xin Miao
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Tao Gao
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Jinlong Suo
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Bingbo Bao
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Yun Qian
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| | - Xianyou Zheng
- Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, P.R. China
| |
Collapse
|
37
|
Song H, Dong C, Ren J. Simultaneously Monitoring Multiple Autophagic Processes and Assessing Autophagic Flux in Single Cells by In Situ Fluorescence Cross-Correlation Spectroscopy. Anal Chem 2024; 96:6802-6811. [PMID: 38647189 DOI: 10.1021/acs.analchem.4c00725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Autophagy is a widely conserved and multistep cellular catabolic process and maintains cellular homeostasis and normal cellular functions via the degradation of some harmful intracellular components. It was reported that high basal autophagic activity may be closely related to tumorigenesis. So far, the fluorescence imaging technique has been widely used to study autophagic processes, but this method is only suitable for distinguishing autophagosomes and autolysosomes. Simultaneously monitoring multiple autophagic processes remains a significant challenge due to the lack of an efficient detection method. Here, we demonstrated a new method for simultaneously monitoring multiple autophagic processes and assessing autophagic flux in single cells based on in situ fluorescence cross-correlation spectroscopy (FCCS). In this study, microtubule-associated protein 1A/1B-light chain 3B (LC3B) was fused with two tandem fluorescent proteins [mCherry red fluorescent protein (mCherry) and enhanced green fluorescent protein (EGFP)] to achieve the simultaneous labeling and distinguishing of multiple autophagic structures based on the differences in characteristic diffusion time (τD). Furthermore, we proposed a new parameter "delivery efficiency of autophagosome (DEAP)" to assess autophagic flux based on the cross correlation (CC) value. Our results demonstrate that FCCS can efficiently distinguish three autophagic structures, assess the induced autophagic flux, and discriminate different autophagy regulators. Compared with the commonly used fluorescence imaging technique, the resolution of FCCS remains unaffected by Brownian motion and fluorescent monomers in the cytoplasm and is well suitable to distinguishing differently colored autophagic structures and monitoring autophagy.
Collapse
Affiliation(s)
- Haohan Song
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Chaoqing Dong
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Jicun Ren
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| |
Collapse
|
38
|
Huang K, Wang Q, Qu H, Hu X, Niu W, Hultgårdh-Nilsson A, Nilsson J, Liang C, Chen Y. Effect of acidosis on adipose-derived stem cell impairment and gene expression. Regen Ther 2024; 25:331-343. [PMID: 38333090 PMCID: PMC10850859 DOI: 10.1016/j.reth.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/10/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Based on disappointing results of stem cell-based application in clinical trials for patients with critical limb ischemia, we hypothesized that the acidic environment might be the key factor limiting cell survival and function. In the present study, we used microdialysis to determine presence of acidosis and metabolic imbalance in critical ischemia. Moreover, we explored the effect of extracellular acidosis on adipose-derived stem cells (ADSCs) at molecular and transcriptional level. Our data demonstrate that low pH negatively regulates cell proliferation and survival, also, it results in cell cycle arrest, mitochondrial dynamics disorder, DNA damage as well as the impairment of proangiogenic function in a pH-dependent manner. Further transcriptome profiling identified the pivotal signaling pathways and hub genes in response to acidosis. Collectively, these findings provide strong evidences for a critical role of acidosis in ADSCs impairment with ischemic condition and suggest treatments focus on tissue pH balance and acidosis-mediated hub genes may have therapeutic potential in stem cell-based application.
Collapse
Affiliation(s)
- Kun Huang
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai Cardiovascular Institute of Integrative Medicine, 200003 Shanghai, China
| | - Qinqin Wang
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai Cardiovascular Institute of Integrative Medicine, 200003 Shanghai, China
| | - Huilong Qu
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai Cardiovascular Institute of Integrative Medicine, 200003 Shanghai, China
| | - Xinyu Hu
- Institute for Molecules and Materials, Radboud University, Nijmegen 6525 AJ, Netherlands
| | - Wenhao Niu
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai Cardiovascular Institute of Integrative Medicine, 200003 Shanghai, China
| | | | - Jan Nilsson
- Department of Clinical Sciences Malmö, Lund University, 20502 Malmö, Sweden
| | - Chun Liang
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai Cardiovascular Institute of Integrative Medicine, 200003 Shanghai, China
| | - Yihong Chen
- Department of Cardiology, Second Affiliated Hospital of Naval Medical University, Shanghai Cardiovascular Institute of Integrative Medicine, 200003 Shanghai, China
- Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| |
Collapse
|
39
|
Natami M, Gorgzadeh A, Gholipour A, Fatemi SN, Firouzeh N, Zokaei M, Mohammed Ali SH, Kheradjoo H, Sedighi S, Gholizadeh O, Kalavi S. An overview on mRNA-based vaccines to prevent monkeypox infection. J Nanobiotechnology 2024; 22:86. [PMID: 38429829 PMCID: PMC10908150 DOI: 10.1186/s12951-024-02355-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
The human monkeypox virus (Mpox) is classified as a member of the Poxviridae family and belongs to the Orthopoxvirus genus. Mpox possesses double-stranded DNA, and there are two known genetic clades: those originating in West Africa and the Congo Basin, commonly known as Central African clades. Mpox may be treated with either the vaccinia vaccination or the therapeutics. Modifying the smallpox vaccine for treating and preventing Mpox has shown to be beneficial because of the strong link between smallpox and Mpox viruses and their categorization in the same family. Cross-protection against Mpox is effective with two Food and Drug Administration (FDA)-approved smallpox vaccines (ACAM2000 and JYNNEOSTM). However, ACAM2000 has the potential for significant adverse effects, such as cardiac issues, whereas JYNNEOS has a lower risk profile. Moreover, Mpox has managed to resurface, although with modified characteristics, due to the discontinuation and cessation of the smallpox vaccine for 40 years. The safety and efficacy of the two leading mRNA vaccines against SARS-CoV-2 and its many variants have been shown in clinical trials and subsequent data analysis. This first mRNA treatment model involves injecting patients with messenger RNA to produce target proteins and elicit an immunological response. High potency, the possibility of safe administration, low-cost manufacture, and quick development is just a few of the benefits of RNA-based vaccines that pave the way for a viable alternative to conventional vaccines. When protecting against Mpox infection, mRNA vaccines are pretty efficient and may one day replace the present whole-virus vaccines. Therefore, the purpose of this article is to provide a synopsis of the ongoing research, development, and testing of an mRNA vaccine against Mpox.
Collapse
Affiliation(s)
- Mohammad Natami
- Department of Urology, Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Arsalan Gholipour
- Free Researchers, Biotechnology and Nanobiotechnology, Babolsar, Iran
| | | | - Nima Firouzeh
- Vector-borne Diseases Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Maryam Zokaei
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | | | | | - Shaylan Kalavi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Islamic Azad University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Tang S, Geng Y, Wang Y, Lin Q, Yu Y, Li H. The roles of ubiquitination and deubiquitination of NLRP3 inflammasome in inflammation-related diseases: A review. BIOMOLECULES & BIOMEDICINE 2024; 24:708-721. [PMID: 38193803 PMCID: PMC11293225 DOI: 10.17305/bb.2023.9997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024]
Abstract
The inflammatory response is a natural immune response that prevents microbial invasion and repairs damaged tissues. However, excessive inflammatory responses can lead to various inflammation-related diseases, posing a significant threat to human health. The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome is a vital mediator in the activation of the inflammatory cascade. Targeting the hyperactivation of the NLRP3 inflammasome may offer potential strategies for the prevention or treatment of inflammation-related diseases. It has been established that the ubiquitination and deubiquitination modifications of the NLRP3 inflammasome can provide protective effects in inflammation-related diseases. These modifications modulate several pathological processes, including excessive inflammatory responses, pyroptosis, abnormal autophagy, proliferation disorders, and oxidative stress damage. Therefore, this review discusses the regulation of NLRP3 inflammasome activation by ubiquitination and deubiquitination modifications, explores the role of these modifications in inflammation-related diseases, and examines the potential underlying mechanisms.
Collapse
Affiliation(s)
- Shaokai Tang
- School of Physical Education, Yanshan University, Qinhuangdao, China
| | - Yuanwen Geng
- School of Physical Education, Yanshan University, Qinhuangdao, China
| | - Yawei Wang
- School of Public Administration, Yanshan University, Qinhuangdao, China
| | - Qinqin Lin
- School of Physical Education, Yanshan University, Qinhuangdao, China
- School of Public Administration, Yanshan University, Qinhuangdao, China
| | - Yirong Yu
- School of Physical Education, Yanshan University, Qinhuangdao, China
| | - Hao Li
- School of Physical Education, Yanshan University, Qinhuangdao, China
| |
Collapse
|
41
|
Ahmad I, Fatemi SN, Ghaheri M, Rezvani A, Khezri DA, Natami M, Yasamineh S, Gholizadeh O, Bahmanyar Z. An overview of the role of Niemann-pick C1 (NPC1) in viral infections and inhibition of viral infections through NPC1 inhibitor. Cell Commun Signal 2023; 21:352. [PMID: 38098077 PMCID: PMC10722723 DOI: 10.1186/s12964-023-01376-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/01/2023] [Indexed: 12/17/2023] Open
Abstract
Viruses communicate with their hosts through interactions with proteins, lipids, and carbohydrate moieties on the plasma membrane (PM), often resulting in viral absorption via receptor-mediated endocytosis. Many viruses cannot multiply unless the host's cholesterol level remains steady. The large endo/lysosomal membrane protein (MP) Niemann-Pick C1 (NPC1), which is involved in cellular cholesterol transport, is a crucial intracellular receptor for viral infection. NPC1 is a ubiquitous housekeeping protein essential for the controlled cholesterol efflux from lysosomes. Its human absence results in Niemann-Pick type C disease, a deadly lysosomal storage disorder. NPC1 is a crucial viral receptor and an essential host component for filovirus entrance, infection, and pathogenesis. For filovirus entrance, NPC1's cellular function is unnecessary. Furthermore, blocking NPC1 limits the entry and replication of the African swine fever virus by disrupting cholesterol homeostasis. Cell entrance of quasi-enveloped variants of hepatitis A virus and hepatitis E virus has also been linked to NPC1. By controlling cholesterol levels, NPC1 is also necessary for the effective release of reovirus cores into the cytoplasm. Drugs that limit NPC1's activity are effective against several viruses, including SARS-CoV and Type I Feline Coronavirus (F-CoV). These findings reveal NPC1 as a potential therapeutic target for treating viral illnesses and demonstrate its significance for several viral infections. This article provides a synopsis of NPC1's function in viral infections and a review of NPC1 inhibitors that may be used to counteract viral infections. Video Abstract.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - Mohammad Ghaheri
- Student Research Committee, Alborz University of Medical Sciences, Karaj, Iran
| | - Ali Rezvani
- Anesthesiology Department, Case Western Reserve University, Cleveland, USA
| | - Dorsa Azizi Khezri
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Natami
- Department of Urology, Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | | | - Zahra Bahmanyar
- School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
42
|
Abu Bakar NFAB, Yeo ZL, Hussin F, Madhavan P, Lim V, Jemon K, Prabhakaran P. Synergistic effects of combined cisplatin and Clinacanthus nutans extract on triple negative breast cancer cells. J Taibah Univ Med Sci 2023; 18:1220-1236. [PMID: 37250812 PMCID: PMC10209459 DOI: 10.1016/j.jtumed.2023.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/18/2023] [Accepted: 04/05/2023] [Indexed: 05/31/2023] Open
Abstract
Objective Triple negative breast cancer (TNBC) is the most invasive breast cancer subtype enriched with cancer stem cells. TNBCs do not express estrogen, progesterone, or human epidermal growth factor receptor 2 (HER2) receptors, making them difficult to be targeted by existing chemotherapy treatments. In this study, we attempted to identify the effects of combined cisplatin and Clinacanthus nutans treatment on MDA-MD-231 and MDA-MB-468 breast cancer cells, which represent TNBC subtypes. Methods The phytochemical fingerprint of C. nutans ethanolic leaf extract was evaluated by LC-MS/MS analysis. We investigated the effects of cisplatin (0-15.23 μg/mL), C. nutans (0-50 μg/mL), and a combination of cisplatin (3.05 μg/mL) and C. nutans (0-50 μg/mL), on cell viability, proliferation, apoptosis, invasion, mRNA expression in cancer stem cells (CD49f, KLF4), and differentiation markers (TUBA1A, KRT18) in TNBC cells. In addition, we also studied the interaction between cisplatin and C. nutans. Results Derivatives of fatty acids, carboxylic acid ester, and glycosides, were identified as the major bioactive compounds with potential anticancer properties in C. nutans leaf extract. Reductions in cell viability (0-78%) and proliferation (2-77%), as well as a synergistic anticancer effect, were identified in TNBC cells when treated with a combination of cisplatin and C. nutans. Furthermore, apoptotic induction via increased caspase-3/7 activity (MDA-MB-231: 2.73-fold; MDA-MB-468: 3.53-fold), and a reduction in cell invasion capacity to 36%, were detected in TNBC cells when compared to single cisplatin and C. nutans treatments. At the mRNA level, cisplatin and C. nutans differentially regulated specific genes that are responsible for proliferation and differentiation. Conclusion Our findings demonstrate that the combination of cisplatin and C. nutans represents a potential treatment for TNBC.
Collapse
Affiliation(s)
| | - Zhin Leng Yeo
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Malaysia
| | - Faisal Hussin
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Malaysia
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Vuanghao Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Khairunadwa Jemon
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Malaysia
| | - Praseetha Prabhakaran
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Skudai, Malaysia
| |
Collapse
|
43
|
Chen YR, Zhao RT, Xu YF, Ma YJ, Hu SB, Wang XH, Fan BB, Zhou YJ, Huang YB, Robinson N, Liu JP, Liu ZL. Chinese herbal injections in combination with radiotherapy for advanced pancreatic cancer: A systematic review and network meta-analysis. Integr Med Res 2023; 12:101004. [PMID: 38033651 PMCID: PMC10681939 DOI: 10.1016/j.imr.2023.101004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Background Advanced pancreatic cancer (APC) is a fatal disease with limited treatment options. This study aims to evaluate the effectiveness and safety of different Chinese herbal injections (CHIs) as adjuvants for radiotherapy (RT) in APC and compare their treatment potentials using network meta-analysis. Methods We systematically searched three English and four Chinese databases for randomized controlled trials (RCTs) from inception to July 25, 2023. The primary outcome was the objective response rate (ORR). Secondary outcomes included Karnofsky performance status (KPS) score, overall survival (OS), and adverse events (AEs). The treatment potentials of different CHIs were ranked using the surface under the cumulative ranking curve (SUCRA). The Cochrane RoB 2 tool and CINeMA were used for quality assessment and evidence grading. Results Eighteen RCTs involving 1199 patients were included. Five CHIs were evaluated. Compound Kushen injection (CKI) combined with RT significantly improved ORR compared to RT alone (RR 1.49, 95 % CrI 1.21-1.86). Kanglaite (KLT) plus RT (RR 1.58, 95 % CrI 1.20-2.16) and CKI plus RT (RR 1.49, 95 % CrI 1.16-1.95) were associated with improved KPS score compared to radiation monotherapy, with KLT+RT being the highest rank (SUCRA 72.28 %). Regarding AEs, CKI plus RT was the most favorable in reducing the incidence of leukopenia (SUCRA 90.37 %) and nausea/vomiting (SUCRA 85.79 %). Conclusions CKI may be the optimal choice of CHIs to combine with RT for APC as it may improve clinical response, quality of life, and reduce AEs. High-quality trials are necessary to establish a robust body of evidence. Protocol registration PROSPERO, CRD42023396828.
Collapse
Affiliation(s)
- Yun-Ru Chen
- Centre for Evidence-based Chinese Medicine, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ruo-Tong Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi-Fang Xu
- Department of Oncology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yin-Jie Ma
- Wangjing Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Shao-Bo Hu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Xue-Hui Wang
- Centre for Evidence-based Chinese Medicine, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bing-Bing Fan
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan-Ji Zhou
- Health Management Department, Aerospace Center Hospital, Beijing, China
| | - Yu-Bei Huang
- Department of Epidemiology and Biostatistics, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Nicola Robinson
- Centre for Evidence-based Chinese Medicine, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- School of Health and Social Care, London South Bank University, London, UK
| | - Jian-Ping Liu
- Centre for Evidence-based Chinese Medicine, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhao-Lan Liu
- Centre for Evidence-based Chinese Medicine, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
44
|
Sadeghi MS, Lotfi M, Soltani N, Farmani E, Fernandez JHO, Akhlaghitehrani S, Mohammed SH, Yasamineh S, Kalajahi HG, Gholizadeh O. Recent advances on high-efficiency of microRNAs in different types of lung cancer: a comprehensive review. Cancer Cell Int 2023; 23:284. [PMID: 37986065 PMCID: PMC10661689 DOI: 10.1186/s12935-023-03133-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023] Open
Abstract
Carcinoma of the lung is among the most common types of cancer globally. Concerning its histology, it is categorized as a non-small cell carcinoma (NSCLC) and a small cell cancer (SCLC) subtype. MicroRNAs (miRNAs) are a member of non-coding RNA whose nucleotides range from 19 to 25. They are known to be critical regulators of cancer via epigenetic control of oncogenes expression and by regulating tumor suppressor genes. miRNAs have an essential function in a tumorous microenvironment via modulating cancer cell growth, metastasis, angiogenesis, metabolism, and apoptosis. Moreover, a wide range of information produced via several investigations indicates their tumor-suppressing, oncogenic, diagnostic assessment, and predictive marker functions in different types of lung malignancy. miRNA mimics or anti-miRNAs can be transferred into a lung cancer cell, with possible curative implications. As a result, miRNAs hold promise as targets for lung cancer treatment and detection. In this study, we investigate the different functions of various miRNAs in different types of lung malignancy, which have been achieved in recent years that show the lung cancer-associated regulation of miRNAs expression, concerning their function in lung cancer beginning, development, and resistance to chemotherapy, also the probability to utilize miRNAs as predictive biomarkers for therapy reaction.
Collapse
Affiliation(s)
- Mohammad Saleh Sadeghi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Lotfi
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Narges Soltani
- School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Kalantari L, Ghotbabadi ZR, Gholipour A, Ehymayed HM, Najafiyan B, Amirlou P, Yasamineh S, Gholizadeh O, Emtiazi N. A state-of-the-art review on the NRF2 in Hepatitis virus-associated liver cancer. Cell Commun Signal 2023; 21:318. [PMID: 37946175 PMCID: PMC10633941 DOI: 10.1186/s12964-023-01351-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/09/2023] [Indexed: 11/12/2023] Open
Abstract
According to a paper released and submitted to WHO by IARC scientists, there would be 905,700 new cases of liver cancer diagnosed globally in 2020, with 830,200 deaths expected as a direct result. Hepatitis B virus (HBV) hepatitis C virus (HCV), and hepatitis D virus (HDV) all play critical roles in the pathogenesis of hepatocellular carcinoma (HCC), despite the rising prevalence of HCC due to non-infectious causes. Liver cirrhosis and HCC are devastating consequences of HBV and HCV infections, which are widespread worldwide. Associated with a high mortality rate, these infections cause about 1.3 million deaths annually and are the primary cause of HCC globally. In addition to causing insertional mutations due to viral gene integration, epigenetic alterations and inducing chronic immunological dysfunction are all methods by which these viruses turn hepatocytes into cancerous ones. While expanding our knowledge of the illness, identifying these pathways also give possibilities for novel diagnostic and treatment methods. Nuclear factor erythroid 2-related factor 2 (NRF2) activation is gaining popularity as a treatment option for oxidative stress (OS), inflammation, and metabolic abnormalities. Numerous studies have shown that elevated Nrf2 expression is linked to HCC, providing more evidence that Nrf2 is a critical factor in HCC. This aberrant Nrf2 signaling drives cell proliferation, initiates angiogenesis and invasion, and imparts drug resistance. As a result, this master regulator may be a promising treatment target for HCC. In addition, the activation of Nrf2 is a common viral effect that contributes to the pathogenesis, development, and chronicity of virus infection. However, certain viruses suppress Nrf2 activity, which is helpful to the virus in maintaining cellular homeostasis. In this paper, we discussed the influence of Nrf2 deregulation on the viral life cycle and the pathogenesis associated with HBV and HCV. We summed up the mechanisms for the modulation of Nrf2 that are deregulated by these viruses. Moreover, we describe the molecular mechanism by which Nrf2 is modulated in liver cancer, liver cancer stem cells (LCSCs), and liver cancer caused by HBV and HCV. Video Abstract.
Collapse
Affiliation(s)
- Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Arsalan Gholipour
- Nanotechnology Research Institute, School of Chemical Engineering, Babol Noshirvani University of Technology, Babol, Iran
| | | | - Behnam Najafiyan
- Faculty of Pharmacy, Shiraz University of Medical Science, Shiraz, Iran
| | - Parsa Amirlou
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Nikoo Emtiazi
- Department of Pathology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Faghihkhorasani A, Dalvand A, Derafsh E, Tavakoli F, Younis NK, Yasamineh S, Gholizadeh O, Shokri P. The role of oncolytic virotherapy and viral oncogenes in the cancer stem cells: a review of virus in cancer stem cells. Cancer Cell Int 2023; 23:250. [PMID: 37880659 PMCID: PMC10599042 DOI: 10.1186/s12935-023-03099-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 10/13/2023] [Indexed: 10/27/2023] Open
Abstract
Cancer Stem Cells (CSCs) are the main "seeds" for the initiation, growth, metastasis, and recurrence of tumors. According to many studies, several viral infections, including the human papillomaviruses, hepatitis B virus, Epstein-Barr virus, and hepatitis C virus, promote the aggressiveness of cancer by encouraging the development of CSC features. Therefore, a better method for the targeted elimination of CSCs and knowledge of their regulatory mechanisms in human carcinogenesis may lead to the development of a future tool for the management and treatment of cancer. Oncolytic viruses (OVs), which include the herpes virus, adenovirus, vaccinia, and reovirus, are also a new class of cancer therapeutics that have favorable properties such as selective replication in tumor cells, delivery of numerous eukaryotic transgene payloads, induction of immunogenic cell death and promotion of antitumor immunity, as well as a tolerable safety profile that essentially differs from that of other cancer therapeutics. The effects of viral infection on the development of CSCs and the suppression of CSCs by OV therapy were examined in this paper. The purpose of this review is to investigate the dual role of viruses in CSCs (oncolytic virotherapy and viral oncogenes).
Collapse
Affiliation(s)
| | - Alaleh Dalvand
- Tehran Medical Branch, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Ehsan Derafsh
- Department of Basic Medical Science, Windsor University School of Medicine, Brighton's Estate, Cayton, St. Kitts And Nevis
| | - Farnaz Tavakoli
- Nephrology and Transplantation Ward, Shariati Hospital Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saman Yasamineh
- Young Researchers and Elite Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | | | - Pooria Shokri
- Department of Medical Science, Faculty of Medical Science, Arak University of Medical Sciences, Arak, Iran.
| |
Collapse
|
47
|
Martinelli G, Fumagalli M, Piazza S, Maranta N, Genova F, Sperandeo P, Sangiovanni E, Polissi A, Dell’Agli M, De Fabiani E. Investigating the Molecular Mechanisms Underlying Early Response to Inflammation and Helicobacter pylori Infection in Human Gastric Epithelial Cells. Int J Mol Sci 2023; 24:15147. [PMID: 37894827 PMCID: PMC10607124 DOI: 10.3390/ijms242015147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Helicobacter pylori is a leading cause of chronic gastric inflammation, generally associated with gastritis and adenocarcinoma. Activation of the NF-κB pathway mainly contributes to the inflammatory phenotype observed in H. pylori infection in humans and experimental models. Since the gastric epithelium undergoes rapid turnover, inflammation and pathogenicity of H. pylori result from early phase and chronically activated pathways. In the present study we investigated the early host response to H. pylori in non-tumoral human gastric epithelial cells (GES-1). To dissect the pathogen-specific mechanisms we also examined the response to tumor necrosis factor (TNF), a prototypical cytokine. By analyzing the activation state of NF-κB signaling, cytokine expression and secretion, and the transcriptome, we found that the inflammatory response of GES-1 cells to H. pylori and TNF results from activation of multiple pathways and transcription factors, e.g., NF-κB and CCAAT/enhancer-binding proteins (CEBPs). By comparing the transcriptomic profiles, we found that H. pylori infection induces a less potent inflammatory response than TNF but affects gene transcription to a greater extent by specifically inducing transcription factors such as CEBPβ and numerous zinc finger proteins. Our study provides insights on the cellular pathways modulated by H. pylori in non-tumoral human gastric cells unveiling new potential targets.
Collapse
Affiliation(s)
| | | | - Stefano Piazza
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, 20133 Milan, Italy; (G.M.); (M.F.); (N.M.); (F.G.); (P.S.); (E.S.); (A.P.); (M.D.); (E.D.F.)
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Zhou Y, Yu S, Zhang W. NOD-like Receptor Signaling Pathway in Gastrointestinal Inflammatory Diseases and Cancers. Int J Mol Sci 2023; 24:14511. [PMID: 37833958 PMCID: PMC10572711 DOI: 10.3390/ijms241914511] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/15/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) are intracellular proteins with a central role in innate and adaptive immunity. As a member of pattern recognition receptors (PRRs), NLRs sense specific pathogen-associated molecular patterns, trigger numerous signaling pathways and lead to the secretion of various cytokines. In recent years, cumulative studies have revealed the significant impacts of NLRs in gastrointestinal (GI) inflammatory diseases and cancers. Deciphering the role and molecular mechanism of the NLR signaling pathways may provide new opportunities for the development of therapeutic strategies related to GI inflammatory diseases and GI cancers. This review presents the structures and signaling pathways of NLRs, summarizes the recent advances regarding NLR signaling in GI inflammatory diseases and GI cancers and describes comprehensive therapeutic strategies based on this signaling pathway.
Collapse
Affiliation(s)
- Yujie Zhou
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.Z.); (S.Y.)
| | - Songyan Yu
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.Z.); (S.Y.)
| | - Wenyong Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.Z.); (S.Y.)
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
49
|
You J, Zhou L, San X, Li H, Li M, Wang B. NEDD4 Regulated Pyroptosis Occurred from Co-infection between Influenza A Virus and Streptococcus pneumoniae. J Microbiol 2023; 61:777-789. [PMID: 37792248 DOI: 10.1007/s12275-023-00076-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 10/05/2023]
Abstract
Co-infection of respiratory tract viruses and bacteria often result in excess mortality, especially pneumonia caused by influenza viruses and Streptococcus pneumoniae. However, the synergistic mechanisms remain poorly understood. Therefore, it is necessary to develop a clearer understanding of the molecular basis of the interaction between influenza virus and Streptococcus pneumonia. Here, we developed the BALB/c mouse model and the A549 cell model to investigate inflammation and pyroptotic cell death during co-infection. Co-infection significantly activated the NLRP3 inflammasome and induced pyroptotic cell death, correlated with excess mortality. The E3 ubiquitin ligase NEDD4 interacted with both NLRP3 and GSDMD, the executor of pyroptosis. NEDD4 negatively regulated NLRP3 while positively regulating GSDMD, thereby modulating inflammation and pyroptotic cell death. Our findings suggest that NEDD4 may play a crucial role in regulating the GSDMD-mediated pyroptosis signaling pathway. Targeting NEDD4 represents a promising approach to mitigate excess mortality during influenza pandemics by suppressing synergistic inflammation during co-infection of influenza A virus and Streptococcus pneumoniae.
Collapse
Affiliation(s)
- Jiangzhou You
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610000, People's Republic of China
| | - Linlin Zhou
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610000, People's Republic of China
| | - Xudong San
- Reproductive & Women-Children Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, People's Republic of China
| | - Hailing Li
- Department of Biochemistry, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610000, People's Republic of China
| | - Mingyuan Li
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610000, People's Republic of China.
| | - Baoning Wang
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610000, People's Republic of China.
| |
Collapse
|
50
|
Li K, Wei X, Li K, Zhang Q, Zhang J, Wang D, Yang J. Dietary restriction to optimize T cell immunity is an ancient survival strategy conserved in vertebrate evolution. Cell Mol Life Sci 2023; 80:219. [PMID: 37470873 PMCID: PMC11071854 DOI: 10.1007/s00018-023-04865-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Abstract
Recent advances highlight a key role of transient fasting in optimizing immunity of human and mouse. However, it remains unknown whether this strategy is independently acquired by mammals during evolution or instead represents gradually evolved functions common to vertebrates. Using a tilapia model, we report that T cells are the main executors of the response of the immune system to fasting and that dietary restriction bidirectionally modulates T cell immunity. Long-term fasting impaired T cell immunity by inducing intense autophagy, apoptosis, and aberrant inflammation. However, transient dietary restriction triggered moderate autophagy to optimize T cell response by maintaining homeostasis, alleviating inflammation and tissue damage, as well as enhancing T cell activation, proliferation and function. Furthermore, AMPK is the central hub linking fasting and autophagy-controlled T cell immunity in tilapia. Our findings demonstrate that dietary restriction to optimize immunity is an ancient strategy conserved in vertebrate evolution, providing novel perspectives for understanding the adaptive evolution of T cell response.
Collapse
Affiliation(s)
- Kunming Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ding Wang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|